Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH...

258

Transcript of Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH...

Page 1: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …
Page 2: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Pathophysiology of Postoperative

Ileus: from Bench to Bedside

Page 3: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

FFrans Olivier ThePathofysiology of postoperative ileus: from bench to bedsideThesis University of Amsterdam

© 2008 Frans O. The, Amsterdam, the NetherlandsAll rights reserved. No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopy, recording or any information storage and retrievel system, without written permission of the author.

The research discribed in this thesis was carried out by the department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, the Netherlands and was supported by the Technology Foundation STW, applied science division of NWO and the technology program of the ministry of Economic Affairs (NWO-STW, grant nr AKG.5727).

Edited by: R.A. de Leeuw, idEAct®, Amsterdam, the NetherlandsPrinted by: Buijten & Schipperheijn, Amsterdam, the Netherlands

Page 4: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Pathophysiology of Postoperative

Ileus: from Bench to BedsideACADEMISCH PROEFSCHRIFT

ter verkrijging van de graad van doctor aan de Universiteit van Amsterdam op gezag van de Rector Magnificus

prof. dr. D.C. van den Boomten overstaan van een door het college voor promoties ingestelde

commissie, in het openbaar te verdedigen in de Agnietenkapelop dinsdag 5 februari 2008, te 10.00 uur door

Frans Olivier Thegeboren te Groningen

Page 5: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

PPromotiecommissie: Promotor: Prof. dr. G.E.E. Boeckxstaens

Co-promotor: Dr. W.J. de Jonge

Overige leden: Prof. dr. J.C. KalffProf. dr. D. GrundyProf. dr. J.F.W.M. BartelsmanProf. dr. R.M. BuijsProf. dr. M.W. HollmannProf. dr. W.A. BemelmanProf. dr. M.P.M. Burger

Faculteit der Geneeskunde

Page 6: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

VVoor mijn ouders en Willemijn

Page 7: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Chapter 1General Introduction

Chapter 2 Postoperative Ileus Is Maintained by Intestinal Immune

Infiltrates That Activate Inhibitory Neural Pathways in Mice Gastroenterology 2003; 125: 1137-1147

Chapter 3 The ICAM-1 antisense oligonucleotide ISIS-3082 prevents the

development of postoperative ileus in miceBritish Journal of pharmacology 2005; 146: 252-258

Chapter 4 The vagal anti-inflammatory pathway attenuates intestinal

macrophage activation and inflammation by nicotinic acetylcholine receptor mediated activation of Jak-2/Stat-3.

Nature Immunology 2005; 6: 844-851

Chapter 5 Activation of the Cholinergic Anti-Inflammatory Pathway

Ameliorates Postoperative Ileus in MiceGastroenterolgy 2007; 133: 1219-1228

Chapter 6 Central activation of the cholinergic anti-inflammatory path-

way shortens postoperative ileus in miceSubmitted for publication

Chapter 7Mast Cell Degranulation During Abdominal Surgery Initiates

Postoperative Ileus in MiceGastroenterology 2004; 127: 535-545

Chapter 8Intestinal handling induced mast cell activation and

inflammation in human postoperative ileusGut 2008; 57: 33-40

8

16

44

64

94

118

136

166

Tabl

e of

con

tent

s

Page 8: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Chapter 9Mast Cell Stabilization as Treatment of Postoperative Ileus: a Pilot StudySubmitted for publication

Chapter 10Summery and conclusions

Chapter 11samenvatting en conclusiesdankwoordcolour figures

192

216

228

Page 9: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

1

Page 10: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

1Cha

pter

1General introduction and aim

of this thesis

Page 11: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

10

PGeneral introduction and aim of this thesisPostoperative ileus is a transient motility disorder characterized by impaired gastrointestinal

propulsion in the absence of any mechanical obstruction1. Every abdominal surgical

procedure is followed by some degree of hypomotility and gastrointestinal dysfunction2. The

patient endures nausea, vomiting, abdominal cramping and does not tolerate oral food or

fluid intake3. Besides this considerable discomfort experienced by patients, postoperative

ileus is also an important risk factor for complications such as aspiration pneumonia or

wound dehiscence and subsequently prolongs the duration of hospital admission4, 5. In the

US the annual expenses related to post-operative ileus exceed 1 billion dollars, reflecting

its socio-economical impact3.

Post-operative ileus is still considered inevitable2 and preventative therapeutic strategies

are lacking. In addition, (symptomatic) treatment options have barely improved over the

last decade6. In general, patients are deprived from oral food or fluids until first peristalsis

(a surgeon’s symphony) occurs. Upon this first empirical hallmark oral fluids are cautiously

reintroduced followed by gradual extension of oral intake. Nasogastric decompression

introduced by Wagensteen in 19317 was one of the first and only alleviating therapeutic

interventions and is still the most commonly used strategy combined with iv fluids and

nothing by mouth. Unfortunately, this approach only relieves symptoms and does not

shorten let alone prevent post-operative ileus.

Post-surgical disturbances in gastrointestinal propulsion have been described as early as

the late 19th century when Bayliss and Starling discovered that splanchnic denervation

improves contractility of the gut after laparotomy8. Since then, numerous studies have been

conducted attempting to identify the exact pathophysiological mechanism. Most of these

studies have focused on (autonomous) neurogenic and (stress) hormonal factors1, 9. It is

generally believed that opening of the abdominal cavity and manipulation of the intestines

during surgery activates both somatic and visceral nerve fibers triggering inhibitory neural

pathways10, 11. These inhibitory reflexes are now generally thought to be responsible for the

post-surgical delay in gastrointestinal propulsion12-15. As a consequence, many prokinetic

Page 12: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

1G

ener

al In

trodu

ctio

n

11

drugs have been evaluated to stimulate gastrointestinal motor activity and as such to

overcome this neurogenic inhibitory pathway16-20. However, this strategy has proven rather

ineffective in most clinical trials17, 21. Most likely, this approach has failed, as it is indeed

ineffective to step on the gas without removing the brake. Moreover, postoperative ileus

usually lasts several days, a fact that cannot be explained by activation of visceral nerve

fibers during or immediately after surgery alone. Indeed, once the abdomen is closed,

stimulation of mechano- or pain receptors ceases and other mechanisms should come into

play.

Recently Kalff et al. have shown that in rodents, handling of intestinal loops during

abdominal surgery triggers a mild inflammatory response22 This inflammation is restricted

to the muscularis propria and leads to impaired muscle contractility and subsequent

delayed intestinal transit. This reduction in neuromuscular function develops 4 to 6 hours

after surgery and lasts for more than 24 hours in rodents, most likely explaining why post-

operative ileus can last for several days. Postoperative ileus however, is not restricted to

the small intestine but involves the entire gastrointestinal tract2. One possible explanation

could be that this local inflammation triggers neural pathways affecting the entire gut.

Several studies have indeed shown that epidural infusion of anesthetics shortens ileus23

indirectly suggesting the involvement of a spinal inhibitory neural pathway. In chapter 2 we

evaluated this hypothesis in a mouse model of postoperative ileus.

If inflammation induced by surgical handling is indeed an important pathophysiological

mechanism, more insight in the players and mediators involved is crucial for the development

of drugs interfering with this pathway. One of the eminent events in any inflammatory

response is the extravasation of immune cells from the circulation into the targeted area,

i.e the area of the intestine that has been manipulated. One of the first events leading to

extravasation of leukocytes is the upregulation of adhesion molecules, such as Leukocyte

Function-associated Antigen-1 (LFA-1) and InterCellular Adhesion Molecule-1 (ICAM-1) 24,

25. Agents that interfere with this process reduce inflammation and might therefore represent

interesting tools to shorten post-operative ileus. We tested the potency of antibodies and

anti-sense oligonucleotides targeting ICAM-1 to prevent the influx of inflammatory cells

into the manipulated area and as such shorten postoperative ileus (chapter 3).

Page 13: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

12

Although it seems obvious that manipulation of the intestine is the trigger of the inflammatory

response and therefore should be minimized, it still remains crucial to identify the

mechanism leading to the upregulation of ICAM-1 and other adhesion molecules. Kalff et al

demonstrated that manipulation of the intestine leads to activation of resident macrophages,

a key event in the attraction of leukocytes26. Interestingly, Borovikova et al. reported that

the activation of macrophages by endotoxin can be reduced by vagus nerve stimulation

in a sepsis model27. They demonstrated that this effect is mediated by acetylcholine, the

neurotransmitter released by the vagus nerve, interacting with the alpha7 nicotinic receptor

on the macrophage28. Nicotine indeed dampened macrophage activation by LPS in vitro

leading to a reduction in the release of pro-inflammatory cytokines. Especially as the

gastrointestinal tract is under strict control of the vagus nerve, we explored whether the

anti-inflammatory properties of vagus nerve stimulation also apply to the gastrointestinal

tract (chapter 4), and could represent a powerful tool to reduce inflammation induced by

intestinal manipulation. In chapters 4, 5 and 6, we studied the effect of peripheral and

central activation of the vagus nerve and identified the intracellular signal transduction

pathway mediating the anti-inflammatory effect of nicotine receptor activation in the

macrophages.

Although interference with macrophage function is certainly an interesting therapeutic

approach, an even more preferable strategy would be to prevent macrophage activation

during surgery. The exact mechanisms involved are far from elucidated and subject of

ongoing studies, but one of the most likely triggers is undoubtedly the influx of bacteria.

Schwartz et al. indeed showed that intestinal manipulation correlates with a transient barrier

dysfunction which results in fluorescent micro-sphere translocation29. These micro-spheres,

mimicking luminal bacteria, can be found in mesenteric lymph vessels and monocytes

recruited to the handled gut wall. Based on these findings, we reasoned that this brief

increase in intestinal permeability results from mast cell activation. Intense stimulation

of afferent nerve fibers indeed leads to local release of Calcitonin Gene-Related Peptide

(CGRP) and substance P30, mast cell activation31, and attraction of inflammatory cells,

a mechanism known as neurogenic inflammation32. As mast cells play a central role in

this process and are known to increase mucosal permeability33, 34, we investigated their

possible role in postoperative ileus in chapter 7.

Page 14: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

1G

ener

al In

trodu

ctio

n

13

Based on the studies described in Chapters 2 and 7, the insight in the pathogenesis has

increased considerably creating many opportunities to improve the current treatment of

postoperative ileus. It should be emphasized though that these conclusions are based

on animal studies, and therefore not automatically apply to the human situation. For this

reason, we designed a series of studies evaluating our hypothesis in man. In chapter 8 we

focused on mast cell degranulation, pro-inflammatory mediator release and subsequent

neutrophil influx in response to surgical bowel handling. We compared the extent of mast

cell activation and inflammation during a conventional laparotomy with that of a minimal

invasive surgical procedure. In addition, in-vivo intestinal leukocyte recruitment was

visualized using leukocyte-SPECT scans and post-operative recovery was evaluated in

open and minimal invasive surgical patients.

Finally, in chapter 9 we conducted a randomized double-blind proof of principle study

evaluating the role of mast cell stabilization in the treatment of post-operative ileus in

patients. In summary, the present thesis focuses on the pathogenesis of postoperative

ileus, an iatrogenic disorder with a significant morbidity and economic impact. We have

demonstrated that in contrast to earlier believes, postoperative ileus is a local inflammatory

disorder. We identified the cells of the innate immune system that are involved and evaluated

new therapeutic approaches and their mechanism of action. Finally, the animal data were

translated to the human situation and a first step to clinical application was undertaken.

Page 15: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

14

Reference ListLivingston EH, Passaro EP, Jr. Postoperative ileus. Dig.Dis.Sci. 1990;35:121-132.1. Miedema BW, Johnson JO. Methods for decreasing postoperative gut dysmotility. Lancet On-2. col. 2003;4:365-372.Prasad M, Matthews JB. Deflating postoperative ileus. Gastroenterology 1999;117:489-492.3. Collins TC, Daley J, Henderson WH, Khuri SF. Risk factors for prolonged length of stay after 4. major elective surgery. Ann.Surg. 1999;230:251-259.Longo WE, Virgo KS, Johnson FE, Oprian CA, Vernava AM, Wade TP, Phelan MA, Henderson 5. WG, Daley J, Khuri SF. Risk factors for morbidity and mortality after colectomy for colon can-cer. Dis.Colon Rectum 2000;43:83-91.Luckey A, Livingston E, Tache Y. Mechanisms and treatment of postoperative ileus. Arch.Surg. 6. 2003;138:206-214.Wangensteen OH. The Early Diagnosis of Acute Intestinal Obstruction with comments on pa-7. thology and treatment. J.Surg.Obst.& Gyn. 1932;40:1-17.Bayliss WM, Starling EH. The movements and innervations of the small intestine. J.Physiol 8. (Lond). 1899;24:99-143.Person B, Wexner SD. The management of postoperative ileus. Curr.Probl.Surg. 2006;43:6-65.9. De Winter BY, Boeckxstaens GE, De Man JG, Moreels TG, Herman AG, Pelckmans PA. 10. Effect of adrenergic and nitrergic blockade on experimental ileus in rats. Br.J.Pharmacol. 1997;120:464-468.Boeckxstaens GE, Hirsch DP, Kodde A, Moojen TM, Blackshaw A, Tytgat GN, Blommaart PJ. 11. Activation of an adrenergic and vagally-mediated NANC pathway in surgery-induced fundic relaxation in the rat. Neurogastroenterol.Motil. 1999;11:467-474.Bauer AJ, Boeckxstaens GE. Mechanisms of postoperative ileus. Neurogastroenterol.Motil. 12. 2004;16 Suppl 2:54-60.Tache Y, Monnikes H, Bonaz B, Rivier J. Role of CRF in stress-related alterations of gastric 13. and colonic motor function. Ann N Y Acad Sci 1993;697:233-43.Barquist E, Bonaz B, Martinez V, Rivier J, Zinner MJ, Tache Y. Neuronal pathways involved in 14. abdominal surgery-induced gastric ileus in rats. Am.J.Physiol 1996;270:R888-R894.Plourde V, Wong HC, Walsh JH, Raybould HE, Tache Y. CGRP antagonists and capsaicin on 15. celiac ganglia partly prevent postoperative gastric ileus. Peptides 1993;14:1225-1229.Seta ML, Kale-Pradhan PB. Efficacy of metoclopramide in postoperative ileus after exploratory 16. laparotomy. Pharmacotherapy 2001;21:1181-1186.Bonacini M, Quiason S, Reynolds M, Gaddis M, Pemberton B, Smith O. Effect of intravenous 17. erythromycin on postoperative ileus. Am.J.Gastroenterol. 1993;88:208-211.Brown TA, McDonald J, Williard W. A prospective, randomized, double-blinded, placebo-con-18. trolled trial of cisapride after colorectal surgery. Am.J.Surg. 1999;177:399-401.Hallerback B, Bergman B, Bong H, Ekstrom P, Glise H, Lundgren K, Risberg O. Cisapride in 19. the treatment of post-operative ileus. Aliment.Pharmacol.Ther. 1991;5:503-511.Jepsen S, Klaerke A, Nielsen PH, Simonsen O. Negative effect of Metoclopramide in postop-20. erative adynamic ileus. A prospective, randomized, double blind study. Br.J.Surg. 1986;73:290-291.Bungard TJ, Kale-Pradhan PB. Prokinetic agents for the treatment of postoperative ileus in 21. adults: a review of the literature. Pharmacotherapy 1999;19:416-423.Kalff JC, Carlos TM, Schraut WH, Billiar TR, Simmons RL, Bauer AJ. Surgically induced leuko-22. cytic infiltrates within the rat intestinal muscularis mediate postoperative ileus. Gastroenterol-ogy 1999;117:378-387.Kehlet H, Holte K. Review of postoperative ileus. Am.J.Surg. 2001;182:3S-10S.23. Smith CW, Marlin SD, Rothlein R, Toman C, Anderson DC. Cooperative interactions of LFA-1 24. and Mac-1 with intercellular adhesion molecule-1 in facilitating adherence and transendothelial migration of human neutrophils in vitro. J.Clin.Invest 1989;83:2008-2017.Issekutz AC, Rowter D, Springer TA. Role of ICAM-1 and ICAM-2 and alternate CD11/CD18 25.

Page 16: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

1G

ener

al In

trodu

ctio

n

15

ligands in neutrophil transendothelial migration. J.Leukoc.Biol. 1999;65:117-126.Kalff JC, Schraut WH, Simmons RL, Bauer AJ. Surgical manipulation of the gut elicits an intes-26. tinal muscularis inflammatory response resulting in postsurgical ileus. Ann.Surg. 1998;228:652-663.Borovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI, Watkins LR, Wang H, Abumrad N, 27. Eaton JW, Tracey KJ. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 2000;405:458-462.Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S, Li JH, Wang H, Yang H, Ulloa L, 28. Al Abed Y, Czura CJ, Tracey KJ. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature 2003;421:384-388.Schwarz NT, Beer-Stolz D, Simmons RL, Bauer AJ. Pathogenesis of paralytic ileus: intestinal 29. manipulation opens a transient pathway between the intestinal lumen and the leukocytic infil-trate of the jejunal muscularis. Ann.Surg. 2002;235:31-40.Sharkey KA. Substance P and calcitonin gene-related peptide (CGRP) in gastrointestinal 30. inflammation. Ann N Y Acad Sci 1992;664:425-42.Suzuki R, Furuno T, McKay DM, Wolvers D, Teshima R, Nakanishi M, Bienenstock J. Direct 31. neurite-mast cell communication in vitro occurs via the neuropeptide substance P. J.Immunol. 1999;163:2410-2415.Foreman JC. Substance P and calcitonin gene-related peptide: effects on mast cells and in hu-32. man skin. Int Arch Allergy Appl Immunol 1987;82:366-71.Kanwar S, Kubes P. Mast cells contribute to ischemia-reperfusion-induced granulocyte infiltra-33. tion and intestinal dysfunction. Am.J.Physiol 1994;267:G316-G321.Berin MC, Kiliaan AJ, Yang PC, Groot JA, Kitamura Y, Perdue MH. The influence of mast cells 34. on pathways of transepithelial antigen transport in rat intestine. J.Immunol. 1998;161:2561-2566.

Page 17: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

2

Page 18: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

2Cha

pter

2Postoperative Ileus Is Main-

tained by Intestinal Immune

Infiltrates That Activate Inhibi-

tory Neural Pathways in Mice

Wouter J. de Jonge, René M. van den Wijngaard,

Frans O. The, Merel-Linde ter Beek,

Roelof J. Bennink, Guido N. J. Tytgat,

Ruud M. Buijs, Pieter H. Reitsma,

Sander J. van Deventer, Guy E. Boeckxstaens

Gastroenterology 2003; 125: 1137-1147

Page 19: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

18

AbstractBackground & Aims: Postoperative ileus after abdominal surgery largely contributes to

patient morbidity and prolongs hospitalization. We aimed to study its pathophysiology in

a murine model by determining gastric emptying after manipulation of the small intestine.

Methods: Gastric emptying was determined at 6, 12, 24, and 48 hours after abdominal

surgery by using scintigraphic imaging. Intestinal or gastric inflammation was assessed

by immune-histochemical staining and measurement of tissue myeloperoxidase activity.

Neuromuscular function of gastric and intestinal muscle strips was determined in organ

baths. Results: Intestinal manipulation resulted in delayed gastric emptying up to 48 hours

after surgery; gastric half-emptying time 24 hours after surgery increased from 16.0 ± 4.4

minutes after control laparotomy to 35.6 ± 5.4 minutes after intestinal manipulation. The

sustained delay in gastric emptying was associated with the appearance of leukocyte

infiltrates in the muscularis of the manipulated intestine, but not in untouched stomach or

colon. The delay in postoperative gastric emptying was prevented by inhibition of intestinal

leukocyte recruitment. In addition, postoperative neural blockade with hexamethonium (1

mg/kg intraperitoneally) or guanethidine (50 mg/kg intraperitoneally) normalized gastric

emptying without affecting small-intestinal transit. The appearance of intestinal infiltrates after

intestinal manipulation was associated with increased c-fos protein expression in sensory

neurons in the lumbar spinal cord. Conclusions: Sustained postoperative gastroparesis

after intestinal manipulation is mediated by an inhibitory enterogastric neural pathway that

is triggered by inflammatory infiltrates recruited to the intestinal muscularis. These findings

show new targets to shorten the duration of postoperative ileus pharmacologically.

Page 20: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

19

Cha

pter

2N

euro

-Imm

une

Inte

ract

ions

Mai

ntai

n Po

stop

erat

ive

Ileus

PBackgroundPostoperative ileus is characterized by a transient hypomotility of the gastrointestinal

tract that occurs after essentially every abdominal operation.1 It is a major contributor to

postoperative discomfort and results in prolonged hospitalization and increased patient

morbidity2 The pathophysiology of postoperative ileus is unclear, and as a result, current

treatment is limited to supportive procedures—such as nasogastric suction, early

postoperative feeding,3,4 and minimal use of opioid analgesics— that are known to intensify

ileus.5,6 Earlier pharmacological means of accelerating postoperative intestinal motility,

for instance, by antiadrenergic7 or cholinergic8 agents or by inhibiting peripheral opioid

effects on gastrointestinal transit,5 have had limited success.4,6,9 Therefore, more insight

into the mechanism mediating postoperative ileus is required for the development of new

pharmacological strategies to treat postoperative ileus.

Most previous experimental animal studies have focused on the pathophysiology of

instant hypomotility during or directly after abdominal surgery.10-13 This early component

of postoperative ileus results from the activation of mechanoreceptors, nociceptors, or

both by bowel manipulation during surgery. The subsequent stimulation of afferent fibers

triggers both spinal and supraspinal reflexes, inhibiting gastrointestinal motility and causing

an acute generalized postoperative ileus.10 However, because mechanical activation

of mechanoreceptors and nociceptors ceases shortly after closure of the wound, this

mechanism cannot explain the prolonged nature of postoperative ileus. In previous reports,

it has been shown that the sustained phase of postoperative intestinal hypomotility due to

bowel handling results from inflammarory, rather than neuronal, mechanisms.14 Previously,

it has been shown that intestinal handling during abdominal surgery led to an impaired in

vitro contractility and a delayed transit of the manipulated small intestine. The latter resulted

from activation of resident macrophages and the subsequent establishment of a neutrophilic

infiltrate in the muscularis of the small intestine after bowel handling.14 Although this

phenomenon can account for the impaired propulsive motility of the small intestine, it does

not explain the hypomotility of the entire gastrointestinal tract, as observed in postoperative

ileus.15 It should also be emphasized that in human postoperative ileus, small-intestinal

motility recovers within 12 hours after surgery, whereas gastric and colonic motility remain

Page 21: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

20

disturbed for 3–5 days.1,6,15 Therefore, mechanisms other than local inflammation determine

the long-term hypomotility of untouched parts of the gastrointestinal tract.

In this study, our aim was to show in a murine model for postoperative ileus that leukocyte

infiltrates recruited in the intestinal muscularis by selective small-intestinal manipulation

affect the motility of parts of the gastrointestinal tract, distant from the site of manipulation,

by triggering an inhibitory neural pathway.

Page 22: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

21

Cha

pter

2N

euro

-Imm

une

Inte

ract

ions

Mai

ntai

n Po

stop

erat

ive

Ileus

Materials and MethodsAnimalsMice (female BALB/c; Harlan Nederland, Horst, The Netherlands) were kept under

environmentally controlled conditions (lights on from 8:00 AM to 8:00 PM; water and rodent

nonpurified diet ad libitum; 20°C–22°C; 55% humidity). Mice were used at 8–12 weeks of

age. Animal experiments were performed in accordance with the guidelines of the Ethical

Animal Research Committee of the University of Amsterdam.

Surgical ProceduresMice were used at 6–10 weeks of age. After an overnight fast, mice were anesthetized

by an intraperitoneal (IP) injection of a mixture of ketamine (100 mg/kg) and xylazine

(20 mg/kg). Surgery was performed under sterile conditions. Mice (10–12 per treatment

group) underwent control surgery of only laparotomy or of laparotomy followed by intestinal

manipulation. The surgery was performed as follows. A midline abdominal incision was

made, and the peritoneum was opened over the linea alba. The small bowel was carefully

exteriorized, layered on a sterile moist gauze pad, and manipulated from the distal duodenum

to the cecum for 5 minutes by using sterile moist cotton applicators. Contact or stretch on

the stomach or colon was strictly avoided. After the surgical procedure, the abdomen was

closed by a continuous 2-layer suture (Mersilene 6-0 silk; Ethicon, Somerville, NJ). After

closure, mice were allowed to recover for 4 hours in a heated (32°C) recovery cage. After

4 hours, mice were completely recovered from anesthesia. At 6, 12, 24, and 48 hours after

surgery, the gastric emptying rate was measured with gastric scintigraphy (see below).

Thereafter, mice were quickly anesthetized and killed by cervical dislocation, and the

stomach and small intestine were removed for histological analysis.

TreatmentsMonoclonal antibodies against intracellular cell adhesion molecule-1 (anti-CD54 [ICAM-1];

immunoglobulin [Ig]G2b; clone YN1/1.7; 4.5 mg/kg)16 and lymphocyte function–associated

antigen-1 (CD11a [LFA-1]; IgG2a;H154.163; 2.3 mg/kg)16 were dissolved in dialyzed saline

(0.9% sodium chloride) and given by IP injection 1 hour before surgery. Identical quantities

Page 23: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

22

of nonspecific isotypematched IgGs were administered as controls. Hexamethonium (1

mg/kg) or guanethidine (50 mg/kg) was dissolved in sterile 0.9% sodium chloride and

administered by a single IP injection. Hexamethonium was administered 10 minutes, and

guanethidine 1 hour before the onset of gastric emptying tests.

Gastric Emptying and Transit To determine the gastric emptying rate of a noncaloric semiliquid test meal, mice were

orally administered 0.1 mL of a 30 mg/ml methylcellulose solution containing 10 MBq of

technetium-99m (99mTc)-Albures (Nycomed-Amersham, Eindhoven, The Netherlands)

(albumin microcolloid) in water. Caloric solid test meals were prepared by baking 4 mL

of egg yolk mixed with 1 mL of water containing 400 MBq of 99mTc-Albures. Mice were

offered 100 mg of the baked egg yolk, which was consumed within 1 minute. Immediately

after the administration (semiliquid) or consumption (solid) of the test meal, mice were

scanned with a gamma camera set at 140 keV with 20% energy windows, fitted with a

pinhole collimator equipped with a 3-mm tungsten insert. A series of static images of the

entire abdominal region were obtained by scanning for 30 seconds at 16-minute intervals.

Static images were obtained at 1, 16, 32, 48, 64, 80, 96 (semiliquid), and 112 minutes

(solid) after administration of the test meal. The scanning frequency applied (once every 16

minutes) elicited no delay in gastric emptying because of handling stress.17 Static images

were analyzed by using Hermes computer software (Hermes, Stockholm, Sweden). To

determine the gastric emptying rate, a region of interest (ROI) was drawn around the gastric

and total abdominal region in each image obtained. Gastric emptying was measured by

determining the percentage of activity present in the gastric ROI, compared with the total

abdominal ROI, for each image. Subsequently, the gastric half-emptying time (t1⁄2) and

gastric retention at 64 minutes (Ret64) were determined for each individual mouse by

using DataFit software (version 6.1; Oakdale Engineering, Oakdale, PA). To this end, the

modified power exponential function y(t) - 1 - (1 - ekt)b was used, where y(t) is the fractional

meal retention at time t, k is the gastric emptying rate in minutes, and b is the extrapolated

y-intercept from the terminal portion of the curve. For determination of gastrointestinal

transit at 24 hours after surgery, animals were killed at 80 minutes after consumption of

the solid test meal. The abdomen was opened and the stomach clamped. Stomach, small

intestine, cecum, and colon were carefully exteriorized, and small intestine was divided into

Page 24: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

23

Cha

pter

2N

euro

-Imm

une

Inte

ract

ions

Mai

ntai

n Po

stop

erat

ive

Ileus

6 fragments of equal length. The amount of 99mTc present in the stomach, small-intestinal

fragments, cecum, and colon was subsequently counted in a gamma counter. The geometric

center was calculated from each experimental group according to the following formula:

∑(% radioactivity per segment x segment number)/100

Immunohistochemistry Immunohistochemistry was performed as follows: after rehydration, endogenous peroxidase

activity was eliminated by incubating sections in 150 mmol/L of sodium chloride, pH

7.4, and 50% methanol, containing 3% (wt/vol) H2O2. Nonspecific protein-binding sites

were blocked by incubation for 30 minutes in TENG-T buffer (10 mmol/L Tris, 5 mmol/L

ethylenediaminetetraacetic acid [EDTA], 150 mmol/L sodium chloride, 0.25% gelatin, and

0.05% Tween-20, pH 8.0). Serial sections were incubated overnight with an appropriate

dilution of rat monoclonal antibodies raised against LFA-1, CD3, and CD4. Binding of the

primary antibodies was visualized with 3-amino-9-ethyl carbazole (Sigma, St. Louis, MO)

as a substrate, dissolved in sodium acetate buffer (pH 5.0) to which 0.01% H2O2 was

added.

C-fos immunohistochemistry was performed according to Bonaz et al.,18 with modifications.

Mice were anesthetized with a mixture of fentanyl citrate/fluanisone (Hypnorm; Janssen,

Beerse, Belgium) and midazolam (Dormicum; Roche, Mijdrecht, The Netherlands) at either

90 minutes or 24 hours after surgery. Mice were then transcardially perfused (1.6 mL/min)

with 8 mL of a 0.9% NaCl solution, followed by 50 mL of 4% paraformaldehyde in phosphate

buffer (0.1 mol/L; pH 7.4). After perfusion, the spinal cord was rapidly removed, postfixed

overnight in the same fixative at 4°C, and cryoprotected for 24 hours in 30% sucrose

solution containing 0.05% sodium azide. After fixation, part of the lumbar spinal cord (L1

to L6) was embedded in Tissue-Tek (Sakura Finetek Inc., Torrance, CA). Fortymicrometer

transversal sections were cryostat-cut, and freefloating sections were incubated overnight

at 4°C with the primary polyclonal sheep antibody (0.3 μg/mL; Sigma Genosys, St.

Louis, MO) in 0.25% gelatin and 0.5% Triton X-100 in Tris-buffered saline (TBS; pH 7.4).

Sections were washed in TBS and incubated with biotinylated anti-sheep antiserum (Vector

Laboratories, Burlingame, CA) for 1.5 hours at room temperature. After washing in TBS,

Page 25: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

24

sections were processed for avidin– biotin–peroxidase (Vectorstain; Vector Laboratories),

and peroxidase was visualized by using diaminobenzidine in 0.02% nickel sulphate in TBS

as the chromogen. For quantification of the number of c-fos–expressing neurons, positive

nuclei in 30 sections were counted per lumbar spinal cord analyzed (n = 3 per treatment

group).

Muscularis Whole-Mount Preparation Whole mounts of ileal segments were prepared as previously described,14

with slight modifications. In short, ileal segments (1–6 cm distal from the cecum) were

quickly excised, and mesentery was removed. Intestinal segments were cut open along the

mesentery border, fecal content was washed out in ice-cold phosphate-buffered saline, and

segments were pinned flat in a glass dish filled with preoxygenated Krebs–Ringer solution

(pH 7.4). Mucosa was removed, and the remaining full-thickness sheet of muscularis

externa was fixed for 10 minutes in 100% ethanol. Muscularis preparations were stored in

70% ethanol at 4°C until analysis.

Myeloperoxidase Activity AssayTissue myeloperoxidase (MPO) activity was determined as follows: either full-thickness ileal

segments or isolated ileal muscularis was blotted dry, weighed, and homogenized in a 20x

volume of a 20 mmol/L potassium phosphate buffer (pH 7.4). The suspension was centrifuged

(8000g for 20 minutes at 4°C), and the pellet was taken up in 1 mL of a 50 mmol/L potassium

phosphate buffer (pH 6.0) containing 0.5% hexadecyltrimethylammoniumbromide and 10

mmol/L EDTA and stored in 0.1-mL aliquots at -70°C until analysis. Fifty microliters of the

appropriate dilutions of the tissue homogenate was added to 445 µL of assay mixture,

which contained 0.2 mg/mL tetramethylbenzidine in 50 mg of potassium phosphate buffer

(pH 6.0), 0.5% hexadecyltrimethylammoniumbromide, and 10 mmol/L EDTA. The reaction

was started by adding 5 µL of 30 mmol/L H2O2 to the assay mixture, and the mixture was

incubated for 3 minutes at 37°C. After 3 minutes, 30 L of a 300 µg/mL catalase solution

was added to each tube, and tubes were placed on ice for 3 minutes. The reaction was

ended by adding 2 mL of 0.2 mol/L glacial acetic acid and incubating at 37°C for 3 minutes.

Absorbance was read at 655 nm. One unit of MPO activity was defined as the quantity of

MPO activity required to convert 1 µmol of H2O2 to H2O per minute at 25°C by using purified

MPO activity as a standard (Sigma), and activity was given in units per gram of tissue.

Page 26: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

25

Cha

pter

2N

euro

-Imm

une

Inte

ract

ions

Mai

ntai

n Po

stop

erat

ive

Ileus

In Vitro Contractility MeasurementsStomach and ileum were quickly excised and cut open, and fecal content was flushed with

ice-cold Krebs–Ringer solution (pH 7.4). Tissues were pinned down flat on a dissecting

dish. After removal of the mucosa, longitudinal muscle strips (approximately 10 x 5 mm) of

the gastric fundus and antrum, circular muscle strips (approximately 0.7 x 5 mm) from the

antrum, and circular muscle strips of the ileum (approximately 1.0 x 5.0 mm) were mounted

in organ baths (25 mL) filled with Krebs–Ringer solution (pH 7.4), maintained at 37°C,

and continuously aerated with a mixture of 5% CO2 and 95% oxygen. One end of each

muscle strip was anchored to a glass rod and placed between 2 platinum electrodes. The

other end was connected to a strain gauge transducer (type GM2/GM3; Scaime, Juvigny,

France) for continuous recording of isometric tension. Recording and analysis of muscle

contractions were performed with Acknowledge software (Biopac Systems Inc., Goleta,

CA). The gastric and ileal muscle strips were brought to their optimal point of length-tension

relationship by using 3 µmol/L acetylcholine and were then allowed to equilibrate for at

least 60 minutes before experimentation. Neurally mediated contractions of the muscle

strips of both the gastric fundus and the antrum were induced by means of electrical field

stimulation (0.5–16 Hz; 1- and 2-ms pulse duration; 10-second pulse trains). Responses

were always measured at the top of the contractile peak. In a second series of experiments,

contractions were evoked by the muscarinic receptor agonist carbachol (0.1 nmol/L to 3

µmol/L) and prostaglandin F2α (0.1 nmol/L to 3 µmol/L). Between the responses to the

different contractile receptor agonists, tissues were washed 4 times with an interval of

15 minutes. At the end of each experiment, muscle strips were blotted dry and weighed.

Contractions were calculated in grams of contraction per gram of tissue dry weight.

Drugs and SolutionsAcetylcholine, carbachol, prostaglandin F2α, hexamethonium, and guanethidine were

obtained from Sigma. A Krebs–Ringer solution was used that contained 118.3 mmol/L

NaCl, 4.7 mmol/L KCl, 1.2 mmol/L MgSO4 , 1.2 mmol/L KH2PO4 , 2.5 mmol/L CaCl2 ,

25 mmol/L NaHCO3 , 0.026 mmol/L EDTA, and 11.1 mmol/L glucose. Dr Y. van Kooyk,

Free University Amsterdam, kindly provided antibodies against ICAM-1 and LFA-1. Rat

monoclonal antibodies against CD3ε, CD4, and LFA-1 were purchased from Phar-Mingen

(San Diego, CA).

Page 27: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

26

ResultsIntestinal Manipulation Generates a Sustained GastroparesisAt 6, 12, 24, and 48 hours after laparotomy or laparotomy combined with intestinal

manipulation, gastric emptying of a noncaloric semiliquid test meal was measured by

scintigraphic imaging. Examples of such an abdominal scan series of mice that underwent

laparotomy intestinal manipulation are presented in Figure 1. The anesthetics used during

abdominal surgery (ketamine 100 mg/kg and xylazine 20 mg/kg) did not alter postoperative

(>6 hours) gastric emptying.17 Also, as shown in Figure 1B and C, laparotomy alone had no

effect on the rate of gastric emptying at any time after surgery. After intestinal manipulation,

however, gastric emptying was significantly delayed (Figure 1). The delay was especially

pronounced 6 hours after surgery; intestinal manipulation increased Ret64 by 2.5-fold

compared with laparotomy only (Figure 1B). The (t1⁄2) was increased 3-fold (Figure 1B).

Gastric emptying after intestinal manipulation remained significantly delayed at 12 and 24

hours after surgery (Figure 1B), although the animals were fully recovered from surgery

at these time points. At 48 hours after surgery, Ret64 and t1⁄2 in intestinal manipulation–

treated mice had recovered to normal (Figure 1B). Similar results were obtained by using

a caloric solid test meal (Figure 1C). At 24 hours after surgery, gastric emptying of a caloric

solid test meal was delayed to an extent similar to that of the semiliquid test meal: intestinal

manipulation increased the t1⁄2 2.5-fold compared with laparotomy (Figure 1C).

st

st

AL

IMt=0 t=16 t=32 t=48 t=64 t=80 min

Page 28: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

27

Cha

pter

2N

euro

-Imm

une

Inte

ract

ions

Mai

ntai

n Po

stop

erat

ive

Ileus

0

20

40

60

80

0 10 20 30 40 50time after surgery (hrs)

IM t½ (min)IM Ret64(%)

L T½ (min)L Ret64(%)

*

*

*

*

*

*

B

IM T

½ (m

in)

IM R

et64

(%) /

10

30

L IM

20

60

T½ li

quid

(min

)

T½ s

olid

(min

)p<0.

05p<

0.05

C

Figure 1. Gastric emptying is delayed after abdominal surgery. (A) A representative series of planar scinti-graphic scans of mice that underwent laparotomy (L) or intestinal manipulation (IM) is shown. The position of the stomach is indicated (st) with a dotted circle. From these scans, gastric emptying could be repetitively as-sessed for each mouse individually by determining the amount of radioactivity present in the gastric region compared with the total abdominal region. Note the difference in radioactivity in the intestinal region be-tween L and IM mice (arrows) at t=80 minutes. (B) Half-emptying time (t1⁄2; open symbols) and gastric retention after 64 minutes (Ret64; filled symbols) as a function of time after L (squares) or IM (circles). Intes-tinal manipulation, performed at t =0 hours, resulted in a significant (P <0.05) increase in t1⁄2 and Ret64 com-pared with laparotomy at t = 6, 12, and 24 hours after surgery. Similar results were obtained with use of a caloric, solid test meal; t1⁄2 was significantly increased after intestinal manipulation compared with mice that underwent L only (C). *Significant difference from L with 1-way analysis of variance, followed by Dunnett’s multiple comparison test. Data represent mean SEM of 8–15 mice.

Page 29: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

28

Intestinal Manipulation Recruits Leukocytes Into Intestinal Muscularis The delayed gastric emptying at 12, 24, and 48 hours after intestinal manipulation coincided

with an enhanced activity of the neutrophil indicator MPO in transmural ileal homogenates

(Figure 2). At 24 and 48 hours after surgery, intestinal manipulation, but not laparotomy

alone, resulted in a significant (P <0.05) increase in MPO activity measured in homogenates

of ileal tissue (Figure 2) or in ileal homogenates from which the mucosa was stripped off

(Figure 3). No increase in MPO activity was observed at earlier time points after surgery

(Figure 2). Histological analysis of transverse sections of ileal tissue indeed showed the

presence of LFA-1+ leukocytes in the ileal muscularis 24 hours after intestinal manipulation

(Figure 4B), but not after laparotomy alone (Figure 4A). Further immunohistochemical

staining showed that these leukocytes were MPO+, but CD3- and CD4- (data not shown).

Examination of the presence of inflammatory cells containing MPO activity in whole-mount

preparations (Figure 4C–F) and in isolated ileal muscularis tissue (Figure 3) confirmed the

presence of leukocyte infiltrates in the muscularis of manipulated ileum only (Figure 4C and

D). It is important to note that no increased presence of LFA-1+ leukocytes was found in

the muscularis of gastric antrum (Figure 4G and H) or in colonic tissue (data not shown) at

any time point after surgery.

Figure 2. Ileal myeloperoxidase (MPO) activity was selectively in-creased at 12, 24, and 48 hours after surgery with intestinal manip-ulation (IM). MPO activity was de-termined in whole homogenates of ileum isolated 6, 12, 24, and 48 hours after surgery as indicated. MPO activity was significantly in-creased 12, 24, and 48 hours af-ter laparotomy with IM (gray bars) compared with laparotomy only (L; white bars). *Significant difference from L for each time point with a Student t test (P< 0.05). Data rep-resent mean SEM of 6–8 mice.

0

8

16

6 12 24 48

LIM

hrs PO

MPO

act

ivity

(U/g

ilea

l tis

sue)

* p<0.05

* p<0.05

* p<0.05

Page 30: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

29

Cha

pter

2N

euro

-Imm

une

Inte

ract

ions

Mai

ntai

n Po

stop

erat

ive

Ileus

Occurrence of Delayed Gastroparesis Depends on Intestinal Leukocyte InfluxTo evaluate the role of the small-intestinal infiltrate in the development of gastroparesis,

intestinal manipulation mice received a preoperative bolus with monoclonal blocking

antibodies against ICAM-1 and LFA-1 to prevent leukocyte recruitment during the

postoperative period. Analysis of MPO-containing leukocytes in ileal muscularis (Figure

4E) or MPO activity in ileal muscularis homogenates (Figure 3) at 24 hours after intestinal

manipulation showed that antibody treatment inhibited the leukocyte recruitment down to

30% (P <0.05) of untreated ileal segments. Prevention of the postoperative inflammatory

infiltrate did not affect the delay in gastric emptying 6 hours after surgery but normalized

gastric emptying 24 hours after intestinal manipulation (Figure 5). This effect was seen

with a noncaloric liquid, as well as with a caloric solid test meal (Figure 5B). Treatment with

identical quantities of isotype-matched control IgG did not affect leukocyte recruitment or

the observed postoperative delay in gastric emptying. These observations show that the

later phase of postoperative gastric ileus is mediated by an intestinal inflammatory infiltrate.

The antibody regimen could not prevent gastroparesis 6 hours after surgery, which is in line

with the observation that the intestinal MPO activity was not increased at this time point.

0

1

2

3

L IM IM+MAb

* p<0.05

IM+hex

* p<0.05

MPO

act

ivity

(U/g

ilea

l mus

cula

ris)

Figure 3. Intestinal manipulation results in an increase in MPO activity measured in il-eal muscularis. MPO activity was measured in homogenates of ileal muscularis tissue isolated 24 hours after surgery. Laparotomy (L) with intestinal manipulation (IM) was as-sociated with significantly increased MPO activity in ileal muscularis tissue compared with L alone. Treatment with ICAM-1– and LFA-1–blocking antibodies before IM pre-vented the increase in MPO activity (IM_ab). Treatment with hexamethonium did not affect the increased MPO activity found 24 hours after IM (IM+hex). *Significant dif-ference from L with 1-way analysis of vari-ance (P<0.05) followed by Dunnett’s mul-tiple comparison test. Data represent mean SEM of 5–8 mice.

Page 31: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

30

Figure 4. (see fullcolor chapter 11) Focal leukocyte infiltrates after intestinal manipulation in the ileal muscularis tissue. (A and B) Transverse sections of the ileal intestinal muscularis 24 hours after laparotomy (A) and intestinal manipulation (B) were stained with mouse-specific monoclonal rat anti-bodies against LFA-1 (CD11a). Note the presence of LFA-1+ leukocytes in the ileal muscularis after (B) intestinal manipulation (arrows), but not after (A) laparotomy. Sections were counterstained with hematoxylin. MPO activity–containing leukocytes were visualized in whole mounts of ileal muscularis tissue (C–F) isolated 24 hours after surgery. Intestinal manipulation (D), but not laparotomy (C), was associated with a focal influx of MPO-containing leukocytes. Preoperative treatment of the mice with monoclonal rat-blocking antibodies against ICAM-1 (CD54), combined with rat monoclonal antibodies against LFA-1, prevented leukocyte influx (E). Postoperative treatment with hexamethonium did not affect the presence of MPO-staining cells 24 hours after laparotomy with intestinal manipulation (F ). (G and H) Transverse sections of gastric antrum stained with monoclonal antibody against LFA-1. Note the lack of LFA-1_ cells in the antral muscularis after laparotomy (G), as well as laparotomy with intestinal manipulation (H). Sections were counterstained with hematoxylin. Bar is 75 mm (A, B, G, and H) or 0.6 mm (C, D, E, and F ).

Page 32: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

31

Cha

pter

2N

euro

-Imm

une

Inte

ract

ions

Mai

ntai

n Po

stop

erat

ive

Ileus

Postoperative Inflammatory Infiltrates in the Intestinal Muscularis Activate Spinal Afferent Neurons and Result in Gastric IleusNext, we investigated whether the small-intestinal infiltrate induced gastroparesis by

activation of an inhibitory neural pathway. To evaluate afferent neurotransmission in this

context, we measured the induction of the immediate-early gene c-fos within the spinal cord

24 hours after laparotomy or laparotomy with intestinal manipulation. Intestinal manipulation

significantly (P < 0.05) increased the number of nuclei expressing c-fos protein in the lumbar

dorsal horn of the spinal cord compared with laparotomy alone (Figure 6A and B). Most

positively labeled nuclei were found in laminae I of the lumbar dorsal horn. Treatment with

neutralizing antibodies against ICAM-1 and LFA-1 before intestinal manipulation prevented

the increase in spinal c-fos expression (Figure 6A and B), showing that intestinal leukocyte

infiltrates mediate spinal afferent activation. Treatment with control IgG antibodies did not

prevent increased c-fos expression after intestinal manipulation.

To further examine whether the sustained phase of delayed gastric emptying after intestinal

manipulation was neurally mediated, mice were treated either with hexamethonium, an

antagonist of nicotinic receptors (1 mg/kg, 10 minutes before gastric scintigraphy), or with

guanethidine, an adrenergic blocker (50 mg/kg, 1 hour before gastric scintigraphy) at 24

hours after abdominal surgery. These treatments did not affect gastric emptying (t1⁄2 or

Ret64) in control mice that underwent control laparotomy (data not shown). Furthermore,

the treatment with hexamethonium (Figures 3 and 4F) or guanethidine (not shown) did not

affect the leukocyte recruitment in the ileal muscularis after intestinal manipulation at 24

hours. After intestinal manipulation, however, treatment with these neural blockers either

partially (6 hours after surgery) or completely (24 hours after surgery) prevented the delay

in gastric emptying, compared with treatment with vehicle control (Figure 5A and B).

Page 33: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

32

rela

tive

gast

ric c

onte

nt (%

)

Time (min)

6 hr PO

20 60 100p<0.05

24 hr PO

20 60 100

p<0.05

10

30

L IM IM+MAb IM+hex IM+gua

20

60

T½ li

quid

(min

)

T½ s

olid

(min

)p<0.

05p<

0.05

100

60

20

IM

B20 60 100 20 60 100

100

60

20

L

p<0.05

p<0.05

IMMAb

IM+hexIM+gua

IM

A

Page 34: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

33

Cha

pter

2N

euro

-Imm

une

Inte

ract

ions

Mai

ntai

n Po

stop

erat

ive

Ileus

Hexamethonium Ameliorates Postoperative Gastric Emptying, But Not Intestinal TransitBecause normalization of gastric emptying could also be secondary to improvement or

acceleration of intestinal transit, we evaluated the effects of hexamethonium on intestinal

transit. Figure 7 shows that, in mice that underwent intestinal manipulation, the radiolabeled

test meal accumulates in the stomach, and that the small-intestinal transit is delayed

compared with control mice that underwent laparotomy. As indicated in Figure 7, intestinal

manipulation and vehicle (saline) treatment led to a significant decrease of the geometric

center (P < 0.05). Postoperative treatment with hexamethonium prevented this surgery-

induced delay in gastric emptying but did not prevent the delay in small-intestinal transit.

Consequently, the geometric center was not different from that in mice that underwent

intestinal manipulation and received saline (Figure 7). The finding that hexamethonium

treatment normalizes gastric emptying even though intestinal transit is still delayed implies

that the delayed gastric emptying is not secondary to a functional obstruction of the small

intestine. To further evaluate the effect of hexamethonium on the delay in intestinal transit

induced by manipulation, we tested the in vitro contractility of intestinal circular muscle

strips. As shown in Figure 8, intestinal manipulation led to an impaired contractile activity of

circular muscle in response to carbachol. The addition of hexamethonium (3 x 10-5 mol/L)

did not reverse the impaired contractile response (Figure 8).

Figure 5. Gastroparesis after intestinal manipulation (IM) is prevented by blocking leukocyte infiltra-tion or neural blockade by hexamethonium or guanethidine treatment. Gastric emptying, determined by scintigraphic imaging of the abdomen after oral administration of a semiliquid noncaloric meal at 6 and 24 hours (A) after IM, was compared with laparotomy alone (L). Values in (A) are given as rela-tive gastric content compared with the total abdominal region. Corresponding t1⁄2 (B) with semiliquid noncaloric (gray bars) and caloric solid (white bars) test meals were significantly (P < 0.05) increased at 6 and 24 hours after IM, compared with L. Preoperative treatment with anti–ICAM-1 and anti–LFA-1 antibodies (IM+MAb) normalized the t1⁄2 of semiliquid and solid test meals (B) at 24 hours after surgery. Postoperative injections of hexamethonium (IM+hex) or guanethidine (IM+gua) normalized t1⁄2 at 6 and 24 hours (B). Values are averages SEM of 8–12 mice per treatment group. Significant differences (P < 0.05), determined by 1-way analysis of variance with treatment groups as variants, are indicated.

Page 35: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

34

L

IM

B

0

10

20

L IM IM + Mab

Mea

ncfo

scou

nt p

er s

ectio

n

* p<0.05

IM + MAb

A

Figure 6. Expression of c-fos in the spinal cord 24 hours after in-testinal manipulation. (A) c-fos–labeled nuclei in the left and right hemispheres of the lumbar dorsal horn of mice 24 hours after control laparotomy (L), intestinal manipu-lation (IM), or IM with pretreatment

with neutralizing antibodies against ICAM-1 and LFA-1 (IM + MAb). Images are representative of 3 mice examined in each group. The number of nuclei labeled per section was significantly increased after IM (B) compared with control. Pretreatment of mice with neutralizing antibodies against ICAM-1 and LFA-1 prevented increased c-fos expression after intestinal manipulation. Significant differences (P < 0.05), determined by 1-way analysis of variance with treatment group as variants, are indicated. Values are averages SEM of 3 mice per treatment group.

Page 36: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

35

Cha

pter

2N

euro

-Imm

une

Inte

ract

ions

Mai

ntai

n Po

stop

erat

ive

Ileus

Neuromuscular Properties of Gastric Fundus and Antrum Are Not Affected by Intestinal ManipulationTo exclude the possibility that the delayed gastric emptying resulted from impaired local

neuromuscular function, the in vitro contractility of isolated muscle strips from gastric

fundus and antrum was investigated in organ baths. In Figure 9, the isomeric contractile

responses to increasing concentrations of the muscarinic receptor agonist carbachol

(0.1 nmol/L to 3 mmol/L) or prostaglandin F2a (0.1 nmol/L to 3 µmol/L) were determined

from longitudinal (Figure 9A and B) or circular (Figure 9C) muscle strips isolated from

gastric fundus (Figure 9A) and antrum (Figure 9B and C). Intestinal manipulation did not

affect the dose-dependent contractile response to stimulation of gastric muscle strips with

prostaglandin F2α or carbachol, compared with mice that underwent laparotomy alone. F 7

stomach 1 2 3 4 5 6 cecum colon

small intestine (fragment nr)

% o

f tot

al ra

dioa

ctiv

ity

10

20

30

40

50L + salineIM+ salineIM+ hexamethonium

GC ± SEM4.2 ± 0.3*2.6 ± 0.33.0 ± 0.3

Figure 7. Postoperative hexamethonium treatment accelerates postoperative gastric emptying, but not intestinal transit. Transit was measured as a percentage distribution of the nonabsorbable 99mTc-Albures (albumin microcolloid) over the gastrointestinal tract after oral intake of a caloric solid test meal. Stomach and 6 equal segments of small bowel, cecum, and colon were isolated 80 minutes after oral ingestion of the caloric test meal (baked egg yolk), and radioactivity was counted in each segment. In mice that underwent intestinal manipulation (IM) and received vehicle (saline) (dark gray bars), the distribution of radioactivity indicates a delayed gastric emptying and an impaired small-intestinal transit time compared with control mice that underwent only laparotomy (L; black bars). The geometric center (GC) was significantly lower (P < 0.05; 1-way analysis of variance) in mice that received IM + saline. Postoperative treatment with hexamethonium prevented the surgery-induced delay in gastric emptying (IM + hexamethonium; light gray bars), but not intestinal transit. Consequently, the geometric center was not different from that in mice that underwent IM + saline. The impaired intestinal transit after manipulation is highlighted by a higher percentage of radioactiv-ity found in intestinal fragments 1 and 2 in manipulated intestine compared with L and by the lower percentage of radioactivity in fragments 5 and 6 (indicated by the dotted boxes). Numbers shown are averages SEM of 8 mice per group.

Page 37: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

36

In addition, contractions evoked by nerve stimulation (0.5–16 Hz; 1-ms pulse duration; 10-

second pulse trains) in gastric fundus (Figure 9A) and antrum (Figure 9B and C) from mice

that underwent intestinal manipulation were not significantly different from contractions in

those that underwent control laparotomy.

9 8 7 60

0.04

0.08

0.12

cont

ract

ion

(g/g

tiss

ue/m

m2 )

-log[carbachol]

0

0.04

0.08

0.12

9 8 7 6

-log[carbachol]

***

***

A

B

+ saline

+ 3*10-5M hexamethonium

Figure 8. Ileal circular muscle carbachol dose–response curves 24 hours after laparotomy or intes-tinal manipulation. (A) intestinal manipulation (open squares) significantly suppresses contraction to higher doses of carbachol compared with laparotomy (open circles). (B) The addition of hexametho-nium (3 x10-5 mol/L) to the organ bath did not reverse the impaired contractility of mice that underwent intestinal manipulation (filled squares). Values are mean SEM of 6 mice. Contractions are expressed in grams of contraction per gram of tissue per square millimeter. *Significant differences (P < 0.05) after unpaired Student t tests.

Page 38: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

37

Cha

pter

2N

euro

-Imm

une

Inte

ract

ions

Mai

ntai

n Po

stop

erat

ive

Ileus

11

21

41

81

161

162

9 8 7 6-log[carbachol]

2

9 8 7 6

LIM

-log[PGF2α]

0

A

9 8 7 6-log[PGF2α]

11

21

41

81

161

162

0

2

9 8 7 6

1

-log[carbachol]

B LIM

0

2

0

2

0

1

2

1 1 1

0

1

2

(Hz)(ms)

(Hz)(ms)

Figure 9. In vitro gastric contractility of mice that underwent intestinal manipulation was not altered. Lack of effect of intestinal manipulation on in vitro contractility of longitudinal muscle strips of gastric fundus (A) and antrum (B) or circular muscle strips of the antrum (C) on different receptor agonists and electric field stimulation is shown. Dose–response curves after electrical pulse stimulation (left), carbachol (middle), or prostaglandin F2α (right) are shown. There was no difference in the neuromo-tor responses of mice that underwent laparotomy (filled symbols) or intestinal manipulation (open symbols). Contractions are expressed in grams of contraction per gram of tissue per square millime-ter. Values shown are means SEM (n= 6 - 7). No significant differences (P < 0.05) were found after 1-way analysis of variance followed by a Dunnett’s multiple comparison test.

Page 39: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

38

DiscussionPostoperative ileus is associated with vomiting, bloating, nausea, and abdominal pain

and contributes considerably to postoperative patient morbidity. In addition, it has a

major economic effect due to prolonged hospitalization and increased costs of health

care. The annual economic cost resulting from the occurrence of postoperative ileus in

the U.S. population has been estimated to be $750,000,000,2 and this may even be a

gross underestimation, because drug costs and indirect costs were not measured. Until

now, treatment of postoperative ileus has been rather disappointing, mainly because

of a lack of pathophysiological insight. Here we provide data clarifying the underlying

mechanisms of the sustained phase of postoperative ileus. First, we confirmed that bowel

manipulation induces the local influx of inflammatory cells. 14 Subsequently, we showed that

the recruitment of this muscular infiltrate is associated with the activation of an inhibitory

adrenergic neural pathway that leads to prolonged postoperative gastroparesis. Our data

suggest that this mechanism is responsible for the generalized hypomotility observed in

postoperative ileus.

Most previous studies have evaluated only the acute effects of abdominal surgery on

gastrointestinal motility.10,11,19,20 However, we show here that, in mice, intestinal manipulation,

but not laparotomy alone, delays gastric emptying up to 48 hours after surgery. Two phases

can be distinguished in the period of postoperative gastric hypomotility: a first acute phase

that is not related to any inflammatory event and a second, later onset, and more sustained

phase that is temporally associated with a leukocyte influx into the intestinal muscularis.

Abundant evidence has been reported indicating that the mechanism underlying the

first, acute phase is a neurally mediated phenomenon: chemical neural blockade with

capsaicin,20,21 hexamethonium,10 or adrenergic antagonists12 reduced the rate of postoperative

ileus in animal models. In addition, surgical procedures that interrupt neural input to the

investigated gastrointestinal region, such as vagotomy or splanchnectomy,10 prevented or

reduced the postoperative hypomotility. Furthermore, studies evaluating neuronal c-fos

expression showed that both spinal and supraspinal pathways synapsing in the brainstem

are activated during abdominal surgery.22 The inhibitory efferent pathways involved have

been shown to be adrenergic and nonadrenergic noncholinergic in nature.10,11,19

Page 40: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

39

Cha

pter

2N

euro

-Imm

une

Inte

ract

ions

Mai

ntai

n Po

stop

erat

ive

Ileus

In this study, we confirmed that the acute phase of postoperative ileus is mediated by a

neural inhibitory mechanism: the nicotinic antagonist hexamethonium and the adrenergic

blocker guanethidine improved the manipulation-induced delayed gastric emptying. The

observation that guanethidine only partially normalized the gastric emptying after intestinal

manipulation is in concert with the involvement of a nonadrenergic mechanism in the

efferent pathway mediating this phenomenon.10,11 These findings clearly indicate that bowel

manipulation activates neural pathways, most likely via activation of mechanoreceptors or

nociceptors. However, mechanisms other than mechanical activation of these receptors

must be involved after closure of the abdomen to explain for the prolonged phase of

postoperative ileus, which lasts up to 24 hours, as observed in this study.

In this respect, Kalff et al.14 previously described that intestinal manipulation initiated

the up-regulation of ICAM-1 and LFA-1 and the subsequent recruitment of leukocytes

into the intestinal muscularis, leading to impaired contractility of circular muscle strips

of jejunum. It was suggested that these functional changes in the intestinal muscularis

resulting from a local inflammatory response were directly responsible for the sustained

paralysis of the gastrointestinal tract. In this study, we showed that the occurrence of an

inflammatory infiltrate was confined to the manipulated small intestine and was absent

in the non-manipulated stomach or colon. In addition, although the in vitro contractility of

ileal circular muscle strips was impaired after intestinal manipulation (compare with Kalff

et al.14), that of gastric muscle strips was unaffected by intestinal manipulation. The latter

finding shows that the delayed gastric emptying 24 hours after intestinal manipulation is not

due to impaired gastric neuromuscular function related to inflammation.

Instead, our results provide evidence that gastric ileus is the result of activation of an

inhibitory adrenergic neural pathway triggered by manipulation-induced leukocyte infiltrates

in the intestinal muscularis. This evidence is based on 2 main findings. First, the neuronal

blockers guanethidine and hexamethonium normalized postoperative gastric emptying.

Second, we confirmed23 that the occurrence of muscular infiltrates was associated with

the activation of c-fos expression in spinal sensory neurons. Furthermore, blockade

of manipulationinduced intestinal leukocyte recruitment by treatment with neutralizing

antibodies against LFA-1 and its main cellular ligand, ICAM-1,24 prevented postoperative

Page 41: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

40

activation of spinal neurons and normalized gastric emptying. These findings indicate that

the activation of t the adrenergic inhibitory pathway is most probably maintained by the

leukocyte infiltrate in the small-intestinal muscularis. The finding that ICAM-1 treatment

did not normalize the delay in gastric emptying 6 hours after surgery further corroborates

this notion, because no infiltrate was yet present at that time. What specific cell population,

leukocyte-derived mediator, or afferent nerve receptor is responsible for the neuro-immune

interaction leading to the activation of the adrenergic pathway remains to be established.

Alternatively, impaired gastric emptying may simply be secondary to stasis of chyme in

the intestine. The intestinal malfunction resulting from the manipulation-induced muscular

inflammation could theoretically back up the emptying of the stomach. However, we

showed that hexamethonium did normalize gastric emptying even though intestinal

transit remained delayed, making this possibility less likely. The independent modulation

of gastric emptying and intestinal transit is in agreement with previous reports.25,26 The

finding that hexamethonium normalized only gastric emptying and not intestinal transit

does not imply that the inhibitory neural input is confined to the stomach. Rather, the delay

in intestinal transit being resistant to hexamethonium can be explained by the local effect of

manipulation-induced muscular inflammation on intestinal motility.14 Indeed, we found that

hexamethonium did not prevent the occurrence of the infiltrate and had no effect on the

impaired in vitro contractility of the manipulated small intestine. To what extent the inhibitory

neural input contributes to the impaired intestinal transit cannot be determined from our

experiments.

Finally, intestinal inflammation could affect gastric motility via enhanced release of circulating

inflammatory mediators from the site of inflammation, such as the cytokines interleukin-

1β, tumor necrosis factor-α, or interleukin-627; prostaglandins28; bradykinin; or mediators

released by activated mast cells that potentially may affect gastric motility. However, in

our current study, hexamethonium or guanethidine administered 24 hours after surgery

could prevent gastroparesis, which implies that neuronal activity, rather than circulating

mediators, determines the delay in gastric emptying.

Several pathophysiological mechanisms may explain the inflammatory events observed in

surgically manipulated bowel tissue. Mechanical manipulation of the bowel during surgery

Page 42: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

41

Cha

pter

2N

euro

-Imm

une

Inte

ract

ions

Mai

ntai

n Po

stop

erat

ive

Ileus

leads to intense activation of nerve fibers in the gut wall. This may result in local release

of substances with potent proinflammatory properties, such as substance P29 or calcitonin

gene-related peptide,29 which can potentially induce neurogenic inflammation.

In addition, recruitment of leukocytes may also be initiated via the release of proinflammatory

mediators by activated resident intestinal muscularis macrophages14 or mast cells. The

latter are known to be activated by neurally released substance P,30 and massive mast

cell activation has been described in response to manipulation of the gut.31 These leads,

together with our current data, suggest that the anti-inflammatory effects of mast cell

stabilization may be instrumental in shortening the duration of postoperative ileus.

We conclude that postoperative ileus is a neurally mediated disorder that consists of

an early phase, which results from the triggering of afferents by activation of mechano-

receptors, nociceptors, or both after bowel manipulation or trauma, and a second,

prolonged, phase, in which an adrenergic inhibitory pathway is triggered by a local infiltrate.

In the rat, incremental degrees of surgical intestinal manipulation and trauma have been

shown to be proportional to the increase in recruitment of leukocyte infiltrates and the

severity of intestinal paralysis.32 This positive correlation may also explain the relation

between the extent, site, and length of intra-abdominal manipulation duration and the

severity of postoperative ileus found in human studies.6 These findings indicate that to

accelerate resumption of postoperative gastrointestinal motility and patient recovery, bowel

manipulation and the consequent recruitment of leukocytes should be kept minimal during

abdominal surgery, i.e., during laparoscopy. However, our study also shows important new

targets in reducing the duration and severity of postoperative ileus pharmacologically by

inhibiting postoperative recruitment of leukocytes to the intestinal wall, for instance, by using

blocking antibodies33 or antisense nucleotides against ICAM-1.34 Shortening postoperative

ileus is clinically and socioeconomically highly desired, and we anticipate that temporal

perioperative prevention of the influx of inflammatory cells may evolve as a new approach

to reduce postoperative patient morbidity.

Page 43: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

42

Reference ListPrasad M, Matthews JB. Deflating postoperative ileus. Gastroenterology 1999;117:489–492.1. Livingston EH, Passaro EP Jr. Postoperative ileus. Dig Dis Sci 1990;35:121–132.2. Bradshaw BG, Liu SS, Thirlby RC. Standardized perioperative care protocols and reduced 3. length of stay after colon surgery. J Am Coll Surg 1998;186:501–506.Steinbrook RA. An opioid antagonist for postoperative ileus. N Engl J Med 2001;345:988–989.4. Taguchi A, Sharma N, Saleem RM, Sessler DI, Carpenter RL, Seyedsadr M, Kurz A. Selective 5. postoperative inhibition of gastrointestinal opioid receptors. N Engl J Med 2001;345:935–940.Holte K, Kehlet H. Postoperative ileus: a preventable event. Br J Surg 2000;87:1480–1493.6. Smith J, Kelly KA, Weinshilboum RM. Pathophysiology of postoperative ileus. Arch Surg 7. 1977;112:203–209.Ruwart MJ, Klepper MS, Rush BD. Carbachol stimulation of gastrointestinal transit in the post-8. operative ileus rat. J Surg Res 1979;26:18–26.Resnick J, Greenwald DA, Brandt LJ. Delayed gastric emptying and postoperative ileus after 9. nongastric abdominal surgery: partII. Am J Gastroenterol 1997;92:934–940. Boeckxstaens GE, Hirsch DP, Kodde A, Moojen TM, Blackshaw A,Tytgat GN, Blommaart PJ. 10. Activation of an adrenergic and vagallymediated NANC pathway in surgery-induced fundic relaxation in the rat. Neurogastroenterol Motil 1999;11:467–474.Boeckxstaens GE, Hollmann M, Heisterkamp SH, Robberecht P, de Jonge WJ, van Den 11. Wijngaard RM, Tytgat GN, Blommaart PJ. Evidence for VIP(1)/PACAP receptors in the afferent pathway mediating surgery-induced fundic relaxation in the rat. Br J Pharmacol 2000;131:705–710.De Winter BY, Boeckxstaens GE, De Man JG, Moreels TG, Herman AG, Pelckmans PA. 12. Effect of adrenergic and nitrergic blockade on experimental ileus in rats. Br J Pharmacol 1997;120:464–468.Kalff JC, Buchholz BM, Eskandari MK, Hierholzer C, Schraut WH, Simmons RL, Bauer AJ. Bi-13. phasic response to gut manipulation and temporal correlation of cellular infiltrates and muscle dysfunction in rat. Surgery 1999;126:498–509.Kalff JC, Carlos TM, Schraut WH, Billiar TR, Simmons RL, Bauer AJ. Surgically induced leuko-14. cytic infiltrates within the rat intestinal muscularis mediate postoperative ileus. Gastroenterol-ogy 1999; 117:378–387.Kehlet H, Holte K. Review of postoperative ileus. Am J Surg 2001;182:S3–S10. 15. Lub M, van Kooyk Y, Figdor CG. Competition between lymphocyte function-associated antigen 16. 1 (CD11a/CD18) and Mac-1 (CD11b/CD18) for binding to intercellular adhesion molecule-1 (CD54). J Leukoc Biol 1996;59:648–655.Bennink RJ, De Jonge WJ, Symonds EL, Van Den Wijngaard RM, Spijkerboer AL, Benninga 17. MA, Boeckxstaens GE. Validation of gastric-emptying scintigraphy of solids and liquids in mice usingdedicated animal pinhole scintigraphy. J Nucl Med 2003;44:1099–1104.Bonaz B, Plourde V, Tache Y. Abdominal surgery induces Fos immunoreactivity in the rat brain. 18. J Comp Neurol 1994;349:212–222.De Winter BY, Robberecht P, Boeckxstaens GE, De Man JG, Moreels TG, Herman AG, Pelck-19. mans PA. Role of VIP1/PACAP receptors in postoperative ileus in rats. Br J Pharmacol 1998; 124:1181–1186.Barquist E, Bonaz B, Martinez V, Rivier J, Zinner MJ, Tache Y. Neuronal pathways involved in 20. abdominal surgery-induced gastric ileus in rats. Am J Physiol 1996;270:R888–R894. Holzer P, Lippe IT, Amann R. Participation of capsaicin-sensitive afferent neurons in gastric mo-21. tor inhibition caused by laparotomy and intraperitoneal acid. Neuroscience 1992;48:715–722.Bonaz B, Tache Y. Corticotropin-releasing factor and systemic capsaicin-sensitive afferents 22. are involved in abdominal surgeryinduced Fos expression in the paraventricular nucleus of the hypothalamus. Brain Res 1997;748:12–20.

Page 44: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

43

Cha

pter

2N

euro

-Imm

une

Inte

ract

ions

Mai

ntai

n Po

stop

erat

ive

Ileus

Kreiss C, Birder LA, Kiss S, VanBibber MM, Bauer AJ. COX-2 dependent inflammation in-23. creases spinal Fos expression during rodent postoperative ileus. Gut 2003;52:527–534.Marlin SD, Springer TA. Purified intercellular adhesion molecule-1 (ICAM-1) is a ligand for 24. lymphocyte function-associated antigen 1 (LFA-1). Cell 1987;51:813–819.Freeman ME, Cheng G, Hocking MP. Role of alpha- and betacalcitonin gene-related peptide in 25. postoperative small bowel ileus. J Gastrointest Surg 1999;3:39–43.Tanila H, Kauppila T, Taira T. Inhibition of intestinal motility and reversal of postlaparotomy ileus 26. by selective alpha 2-adrenergic drugs in the rat. Gastroenterology 1993;104:819–824.Collins SM. The immunomodulation of enteric neuromuscular function: implications for motility 27. and inflammatory disorders. Gastroenterology 1996;111:1683–1699.Schwarz NT, Kalff JC, Turler A, Engel BM, Watkins SC, Billiar TR, Bauer AJ. Prostanoid pro-28. duction via COX-2 as a causative mechanism of rodent postoperative ileus. Gastroenterology 2001; 121:1354–1371.Sharkey KA. Substance P and calcitonin gene-related peptide (CGRP) in gastrointestinal 29. inflammation. Ann N Y Acad Sci 1992; 664:425–442.Suzuki R, Furuno T, McKay DM, Wolvers D, Teshima R, Nakanishi M, Bienenstock J. Direct 30. neurite-mast cell communication in vitro occurs via the neuropeptide substance P. J Immunol 1999;163: 2410–2415.Moriwaki K, Fujii K, Yuge O. Protein exudation induced by manipulation of the intestines and 31. mesentery during laparotomy in rat. A study of the mechanism of “third space” loss. In Vivo 1997; 11:325–327.Kalff JC, Schraut WH, Simmons RL, Bauer AJ. Surgical manipulation of the gut elicits an 32. intestinal muscularis inflammatory response resulting in postsurgical ileus. Ann Surg 1998;228: 652–663.Kavanaugh AF, Schulze-Koops H, Davis LS, Lipsky PE. Repeat treatment of rheumatoid arthri-33. tis patients with a murine antiintercellular adhesion molecule 1 monoclonal antibody. Arthritis Rheum 1997;40:849–853.Bennett CF, Kornbrust D, Henry S, Stecker K, Howard R, Cooper S, Dutson S, Hall W, Jacoby 34. HI. An ICAM-1 antisense oligonucleotide prevents and reverses dextran sulfate sodium-in-duced colitis in mice. J Pharmacol Exp Ther 1997;280:988–1000.

Page 45: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

3

Page 46: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

3Cha

pter

3The ICAM-1 antisense oligo-

nucleotide ISIS-3082 pre-

vents the development of

postoperative ileus in mice

Frans O. The, Wouter J. de Jonge,

Roel J. Bennink, Rene M. van den Wijngaard

Guy E. Boeckxstaens

British Journal of pharmacology 2005; 146: 252-258

Page 47: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

46

AbstractBackground & Aims: Intestinal manipulation (IM) during abdominal surgery triggers the

influx of inflammatory cells, leading to postoperative ileus. Prevention of this local muscle

inflammation, using intercellular adhesion molecule-1 (ICAM-1) and leukocyte function-

associated antigen-1-specific antibodies, has been shown to shorten postoperative ileus.

However, the therapeutic use of antibodies has considerable disadvantages. The aim of the

current study was to evaluate the effect of ISIS-3082, a mouse-specific ICAM-1 antisense

oligonucleotide, on postoperative ileus in mice Methods: Mice underwent a laparotomy

or a laparotomy combined with IM after treatment with ICAM-1 antibodies, 0.1–10 mgkg-1

ISIS-3082, saline or ISIS-8997 (scrambled control antisense oligonucleotides, 1 and 3

mg kg-1). At 24 h after surgery, gastric emptying of a 99mTC labelled semi-liquid meal was

determined using scintigraphy. Intestinal inflammation was assessed by myeloperoxidase

(MPO) activity in ileal muscle whole mounts. Results: IM significantly reduced gastric

emptying compared to laparotomy. Pretreatment with ISIS-3082 (0.1–1 mg kg-1) as well

as ICAM-1 antibodies (10 mg/kg-1), but not ISIS-8997 or saline, improved gastric emptying

in a dose-dependent manner. This effect diminished with higher doses of ISIS-3082 (3–10

mgkg-1). Similarly, ISIS-3082 (0.1–1 mgkg-1) and ICAM-1 antibodies, but not ISIS-8997

or higher doses of ISIS-3082 (3–10mg kg-1), reduced manipulation-induced inflammation.

Immunohistochemistry showed reduction of ICAM-1 expression with ISIS-3082 only.

Conclusion: ISIS-3082 pretreatment prevents postoperative ileus in mice by reduction of

manipulation-induced local intestinal muscle inflammation. Our data suggest that targeting

ICAM-1 using antisense oligonucleotides may represent a new therapeutic approach to the

prevention of postoperative ileus.

Page 48: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

3IC

AM

-1 A

ntise

nse

Olig

onuc

leot

ides

Pre

vent

Pos

tope

rativ

e Ile

us

47

PBackgroundPostoperative ileus is characterised by a generalised hypomotility of the gastrointestinal tract,

and is observed after almost every abdominal surgical procedure1. Although self-limiting,

postoperative ileus is responsible for increased morbidity and prolonged hospitalisation,

leading to extra costs of between 750 million and 1 billion US dollars1, 2. Mainly due to a lack

of pathophysiological insight, treatment is limited to supportive and conservative measures

such as no oral feeding and intravenous (i.v.) fluids3.

Acute studies have convincingly shown that a laparotomy, but especially handling of the

intestine, inhibits gastrointestinal motility by activation of spinal and supraspinal inhibitory

pathways4-9. Recently, it became clear that manipulation of the intestine also triggers

the influx of inflammatory cells. This process becomes prominent several hours after

abdominal surgery and is now accepted to play a crucial role in the prolonged inhibition of

gastrointestinal motility10-12. This local inflammation not only leads to impaired contractility

of the diseased intestinal segment but also triggers an adrenergic inhibitory neural pathway,

explaining the more generalised aspect of postoperative ileus10-13. Leukocyte function-

associated antigen-1 (LFA-1) and its ligand intercellular adhesion molecule-1 (ICAM-1) are

two adhesion molecules that are crucial in the process of transmigration and recruitment of

leukocytes14, 15. ICAM-1, normally only moderately expressed on vascular endothelium, is

strongly upregulated in response to inflammatory stimuli, including intestinal manipulation

(IM)11, 16, 17.

An important role for ICAM-1 in the development of the inflammatory infiltrate mediating

postoperative ileus is suggested by the observation that administration of a combination

of blocking antibodies to LFA-1 and ICAM-1 prior to abdominal surgery prevented the

recruitment of inflammatory cells in manipulated tissue and postoperative ileus11, 12. Although

it has not been studied whether blockade of only one of these adhesion molecules has

a similar effect, these data indicate that ICAM-1 may be an important target to prevent

postoperative ileus. However, the use of antibodies as therapeutic strategy in humans

still has considerable downsides, such as the formation of neutralizing antibodies or the

development of hypersensitivity reactions18,19.

Page 49: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

48

Antisense oligonucleotides are 15–25-base long oligomers designed to hybridise to the

specific mRNA encoding for the target protein. As such, it prevents the translation of

mRNA, thereby downregulating the expression of the respective protein20, 21. ISIS-3082

is a murine ICAM-1-specific antisense oligonucleotide with anti-inflammatory properties

in experimental models of colitis, and a human-specific form, ISIS-2302 (alicaforsen), is

currently being tested in a clinical trial to evaluate this drug as potential new treatment in

patients with inflammatory bowel disease22, 23. In the present study, we investigated the

efficacy of the antisense oligonucleotide ISIS-3082 to shorten postoperative ileus in our

experimental mouse model.

Page 50: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

3IC

AM

-1 A

ntise

nse

Olig

onuc

leot

ides

Pre

vent

Pos

tope

rativ

e Ile

us

49

Materials and MethodsLaboratory AnimalsFemale Balb/C mice (Harlan Nederland, Horst, The Netherlands), 12–15 weeks old, were

kept under environmentally controlled conditions (light on from 08:00 till 20:00 h; water and

rodent nonpurified diet ad libitum; temperature 20-22°C; 55% humidity). All experiments

were performed after approval of the Ethical Animal Research Committee of the University

of Amsterdam and according to their guidelines.

Surgical Procedures: Abdominal SurgeryMice were anaesthetised by intraperitoneal (i.p.) injection of 10 ml/kg of an anaesthetic

solution containing 0.078 mg/ml fentanyl citrate, 2.5 mg/ml fluanisone (Hypnorm;

Janssen, Beerse, Belgium) and 1.25mg/ml midazolam (Dormicum; Roche, Mijdrecht,

The Netherlands). Surgery was performed under sterile conditions. Mice underwent a

laparotomy, or a laparotomy followed by small IM, as described previously12.

In short, a midline incision was made and the peritoneal cavity was opened along the

linea alba. The small intestine was carefully exteriorised from the distal duodenum until the

cecum and gently manipulated for 5 min using sterile moist cotton applicators. Contact or

stretch of stomach or colon was strictly avoided. After repositioning of the intestinal loops,

the abdomen was closed using a two-layer continuous suture (Mercilene Softsilk 6-0). Mice

recovered from surgery in a temperature-controlled cage at 32°C with free access to water,

but not to food. At 24 h after surgery, gastric emptying was measured. Thereafter, mice

were anaesthetised and killed by cervical dislocation. The small intestine was removed,

flushed in ice-cold phosphate-buffered saline (PBS), and snap frozen in liquid nitrogen or

fixed in ethanol for further analysis.

Drug preparation and treatmentICAM-1 antibody (anti-CD54; IgG2b; clone YN1/1.7)24 was kindly provided by Professor Y.

van Kooyk (Department of Molecular Cell Biology & Immunology, VU University Medical

Center, Amsterdam, The Netherlands). Antibodies were dissolved in sterile 0.9% NaCl and

injected i.p. 1 h prior to the surgical intervention in a dose of 10 mg/kg12.

Page 51: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

50

ICAM-1 antisense oligonucleotide (ISIS-3082) and its scrambled control oligonucleotide

(ISIS-8997) were kindly provided by Dr Frank Bennett (ISIS-Pharmaceuticals, Carlsbad, CA,

U.S.A.). The specific sequences of the oligonucleotides used in this study were: ISIS-3082,

50-TGCATCCCCCAGGCCACCAT-30 and ISIS-8997, 50-CAGCCATGGTTCCCCCCAAC-

30. The final concentration of the oligonucleotide was determined using spectrometry

(Nanodrop ND-1000, Nanodrop Technologies Inc., Wilmington, DE, U.S.A.). ISIS-3082,

ISIS-8997 or their vehicle (sterile 0.9% NaCl) was injected subcutaneously (s.c.) once daily

starting 6 days prior to the surgical procedure. As intracellular localisation of the drug is

only achieved after 24 h, the onset of action of antisense oligonucleotide is not instant25.

Therefore, ISIS-3082 or ISIS-8997 was administered by s.c. injection once a day for 6 days

to achieve a steady-state concentration (approximately five half-lives) prior to surgery26.

ISIS-3082 was administered in a pharmacological range of 0.1, 0.3, 1.0, 3.0 or 10mg/kg,

which has been shown to be effective in DSS-colitis27. As the most effective dose of ISIS-

3082 was 1mg/kg, the control oligonucleotide, ISIS-8997, was tested in the same dose, as

well as a higher dose of 3mg/kg.

Measurement of gastric emptyingAs previously described, gastric emptying rate was determined after gavage of a semi-liquid,

noncaloric test meal (0.1 ml of 3% methylcellulose solution containing 10 MegaBecquerel

(MBq) of 99mTc-Albures. Mice were scanned using a gamma camera set at 140 keV28. The

entire abdominal region was scanned for 30 s, immediately and 80 min after gavage. During

the scanning period, mice were conscious and manually restrained. The static images

obtained were analysed using Hermes computer software (Hermes, Stockholm, Sweden).

Gastric retention was calculated by determining the percentage of activity present in the

gastric region of interest compared to the total abdominal region of interest.

Whole-mount preparationIleal segments (4–6 cm proximal of cecum) were quickly excised. The mesentery was

removed from the intestine, which was cut open along its border. Faecal content was

washed out in ice-cold PBS, after which tissue segments were fixed in 100% ethanol for 10

min. Fixed preparations were kept in 70% ethanol at 41°C until further analysis.

Page 52: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

3IC

AM

-1 A

ntise

nse

Olig

onuc

leot

ides

Pre

vent

Pos

tope

rativ

e Ile

us

51

Before final analysis, segments were stretched 1.5 times to their original size and pinned

down on a glass dish filled with 70% ethanol, after which the mucosa was carefully

removed.

Assessment of leukocyte infiltration of the intestinal muscleFixed preparations were rehydrated by incubation in 50% ETOH and PBS, pH 7.4, for 5

min. To visualise MPO-positive cells, preparations were incubated for 10 min with 3-amino-

9-ethyl carbazole (Sigma, St Louis, MO, U.S.A.) as substrate and dissolved in sodium

acetate buffer (pH 5.0), to which 0.01% H2O2 was added12.

ImmunohistochemistryImmunohistochemical staining for ICAM-1 was performed on acetone fixed transverse

ileal segments. Endogenous peroxidase activity was eliminated by incubation of segments

in methanol containing 0.3% H2O2. Nonspecific protein-binding sites were blocked by

incubation in PBS, pH 7.4, containing 10% of normal goat serum for 10 min. Sections were

incubated overnight with biotinylated hamster anti-mouse ICAM-1 antibodies (Pharmingen,

San Diego, CA, U.S.A.) (dilution 1 : 1000). Next, sections were incubated with ABComplex/

HRP (DAKOCytomation, Glostrup, Denmark) for 30 min. HRP was visualised using

SigmaFast DAB (Sigma-Aldrich, St Louis, MO, U.S.A.), incubating 5 min, and contra-

stained with 2% methyl green for 2 min.

Statistical analysisA sample size of eight animals was used for each treatment group. Statistical analysis was

performed using SPSS 12.02 software for Windows. The data were expressed as mean

± s.e.m. Owing to the sample size, data were considered nonparametrically distributed.

The nonparametric Kruskal-Wallis test was used to analyse the cohort of independent

variables. If the difference between the multiple variables was statistically significant, the

Mann–Whitney test was performed to compare the individual treatment groups, identifying

the specific statistical differences. P<0.05 was considered statistically significant.

Page 53: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

52

ResultsEffect of IM on gastric emptying and local intestinal muscle inflammation 24 h after abdominal surgery At 24 h after abdominal surgery, IM resulted in a significant increase of gastric retention

80 min after gavage of a noncaloric test meal, compared to a laparotomy (Figure 1). The

observed delay in gastric emptying after IM coincided with a profound local intestinal muscle

inflammatory cell influx compared to laparotomy (Figure 2).

Effect of ICAM-1 antisense oligonucleotide (ISIS-3082) pretreatment on gastric emptying and intestinal muscle inflammation 24 h after abdominal surgery Pretreatment with ISIS-3082 (0.1–1 mg/kg) reduced gastric retention in a dose-dependent

manner, restoring gastric emptying 24 h after IM at a dosage of 1mg/kg (Figure 3). This

effect was not observed with higher dosages (3– 10 mg/kg) (Figure 3). Moreover, ISIS-

3082 did not affect gastric emptying 24 h after a laparotomy in the absence of IM in mice

treated with 1mg/kg, compared to their vehicle control. In contrast, 1 and 3mg/kg ISIS-

8997, the scrambled control antisense oligonucleotides, did not improve gastric retention

24 h after IM (Figure 3).

10mg/k

g ICAM Ab I

M IML0.0

0.1

0.2

0.3

0.4

0.5

Gas

tric

rete

ntio

n(%

of t

otal

)

*

**

Figure 1 Effect of IM on gas-tric retention 24 h after ab-dominal surgery compared to laparotomy only (L), or IM after treatment with ICAM-1 antibodies (anti-CD54 IgG2b clone 1/1.7). Each individual group consisted of eight ani-mals. Data are mean ± s.e.m. gastric retention 80 min after gavage of semi-liquid test meal; *P<0.05 compared to L control; **P<0.05 compared to IM control.

Page 54: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

3IC

AM

-1 A

ntise

nse

Olig

onuc

leot

ides

Pre

vent

Pos

tope

rativ

e Ile

us

53

The number of MPO-positive cells in muscle whole mounts diminished dose-dependently

(0.1–1 mg/kg) in mice treated with ISIS-3082 (Figure 4). Higher doses (3–10 mg/kg),

however, did not elicit reduction of the cellular infiltrate, nor did the scrambled control

antisense oligonucleotide (1 and 3mg/kg). To evaluate whether administration of high

doses of ISIS-3082 had a local pro-inflammatory effect27, 29, we also studied the effect of

10 mg/kg on animals who only underwent laparotomy. 10 mg/kg of ISIS-3082 did not show

an increase in MPO positive cells after laparotomy (Figure 4). Similar to 1mg/kg ISIS-

3082, administration of ICAM-1-specific antibodies (10 mg/kg i.p.) 1 h before IM resolved

the impaired gastric emptying observed 24 h after surgery, and significantly reduced the

manipulation-induced leukocyte influx (Figures 1, 2, 5a–f).

Small-intestinal ICAM-1 expression Figure 6 shows the immunohistochemical staining for

ICAM-1 on transverse ileal tissue segments to assess the in situ effect.

L IM

0

100

200

300

Inte

stin

al M

uscl

e In

flam

mat

ion

( MPO

-pos

./mm

-2

)

10mg/k

g ICAM Ab I

M

*

**

Figure 2 Effect of IM, laparo-tomy only (L) or IM pretreated with ICAM-1 antibodies (anti-CD54 IgG2b clone 1/1.7) on lo-cal inflammatory cell influx 24 h after abdominal surgery. Each individual group consisted of eight animals. Data are mean ± s.e.m. number of MPO-positive cellsmm-2; *P<0.05 compared to L control; **P<0.05 com-pared to IM control.

Page 55: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

54

Saline L

Saline I

M

0.1mg k

g -1 IM

0.3 m

g kg

-1 IM

1.0 m

g kg

-1 IM

3.0 m

g kg

-1 IM

10.0

mg kg

-1 IM

ISIS-8997

1.0 m

g kg-1 IM

0

10

20

30

Gas

tric

rete

ntio

n(%

of t

otal

)

1 mg k

g -1 L

*

**

ISIS-8997

3.0 m

g kg-1 IM

Figure 3 Effect of ISIS-3082, ISIS-8997 or vehicle on gastric retention 24 h after laparotomy (L) or laparotomy with IM. Each individual group consisted of eight animals. Data are mean ± s.e.m. gas-tric retention 80 min after gavage of semi-liquid test meal; *P<0.05 compared to L control; **P<0.05 compared to vehicle IM control.

Page 56: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

3IC

AM

-1 A

ntise

nse

Olig

onuc

leot

ides

Pre

vent

Pos

tope

rativ

e Ile

us

55

Saline L

Saline I

M

0.1 m

g kg

-1 IM

0.3 m

g kg

-1 IM

1.0 m

g kg

-1 IM

3.0 m

g kg

-1 IM

10.0

mg kg

-1 IM

ISIS-8997

1.0 m

g kg-1 IM

0

100

200

300

Inte

stin

al M

uscl

e In

flam

mat

ion

( MPO

-pos

. mm

-2) *

10.0

mg kg

-1 L

****

ISIS-8997

3.0 m

g kg-1 IM

Figure 4 Effect of ISIS-3082, ISIS-8997 or vehicle on local inflammatory cell influx 24 h after lapa-rotomy (L) or laparotomy with IM. Each individual group consisted of eight animals. Data are mean ± s.e.m. number of MPO-positive cells/mm-2; *P<0.05 compared to L control; **P<0.05 compared to vehicle IM control.

Page 57: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

56

Figure 5 MPO staining of muscle whole mounts from mice that underwent a laparotomy after pre-treatment with saline (a), or a laparotomy with intestinal manipulation after pretreatment with saline (b), ICAM-1 antibodies (10mg/kg) (c), 1mg/kg ISIS-3082 (d), 10 mg/kg ISIS-3082 (e) or 1mg/kg ISIS-8997 (f). Magnification x20; insertion x65.

5a 5b

5c

5f5e

5d

Page 58: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

3IC

AM

-1 A

ntise

nse

Olig

onuc

leot

ides

Pre

vent

Pos

tope

rativ

e Ile

us

57

Figure 6 (see fullcolor chapter 11) ICAM-1 staining of ileal transverse segments from mice pretreated with saline that underwent a laparotomy (a), and from mice that underwent a laparotomy with IM after pretreatment with saline (b), 1mg/kg ISIS-3082 (c), 10 mg/kg ISIS-3082 (d) or 1mg/kg ISIS-8997 (e). Note the increased ICAM-1 expression in the densely vascularised submucosa, but also in the blood vessels, visible in the muscularis propria after IM (arrow heads). Only pretreatment with 1mg/kg ISIS-3082 reduces the ICAM-1 expression (c).

Page 59: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

58

DiscussionIn the present study, we show that both ICAM-1 antibodies and the antisense oligonucleotide

ISIS-3082, targeted against ICAM-1, attenuate postoperative ileus by reducing manipulation-

induced inflammation. These findings illustrate the importance of ICAM-1 in the pathogenesis

of postoperative

ileus, and suggest that ISIS-3082 may represent a potential new pharmacological approach

to prevent postoperative ileus.

Postoperative ileus complicates abdominal surgical intervention and causes prolonged

hospitalisation1. With regard to its pathophysiology, it has been shown that intestinal

handling during abdominal surgery activates mast cells and resident macrophages, initiating

the recruitment of neutrophils into the intestinal muscle layer10-12, 30. This local infiltrate of

leukocytes is now recognised as a crucial player in postoperative ileus, as it has been

shown to activate inhibitory neural pathways that lead to a generalised hypomotility of the

gastrointestinal tract12, 13. Although it is not known to what extent the same mechanism is

responsible for the development of postoperative ileus in patients, Kalff et al.31 observed an

increase in mRNA expression in the human intestine for several proinflammatory proteins

like LFA-1, iNOS, IL-6 and TNF-a after abdominal surgery.

Upregulation of adhesion molecules such as LFA-1 and ICAM-1 are necessary for the

extravasation of leukocytes. Here, we show that ICAM-1 expression is clearly increased

after IM, being most profound in the vasculature between the submucosal and the muscle

layers, but also in the muscularis propria. This observation confirms that manipulation of

the small intestine indeed increases the expression of ICAM-1, facilitating local infiltration of

inflammatory cells 10,11. Previous studies demonstrated that pretreatment with a combination

of antibodies against LFA-1 and ICAM-1 prevented postoperative ileus by blocking of this

manipulation-induced infiltrate12. In the present study, we show that pretreatment with

antibodies targeted to ICAM-1 alone also results in a reduction of inflammatory cell influx

and the prevention of delayed gastric emptying. These results illustrate that ICAM-1 is an

important target to prevent postoperative ileus.

Page 60: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

3IC

AM

-1 A

ntise

nse

Olig

onuc

leot

ides

Pre

vent

Pos

tope

rativ

e Ile

us

59

The use of antisense oligonucleotides is a novel approach to block the synthesis of regulatory

peptides. These 15–25-baselong oligomers hybridise to the specific mRNA, preventing its

translation, thereby downregulating the expression of the respective protein20, 21. ISIS-3082

is a mouse-specific ICAM-1 antisense oligonucleotide, which has been shown to be effective

in experimental murine models for heart allograft rejection and inflammatory bowel disease27,

32. We used ISIS-3082 to study its anti-inflammatory effects in our experimental model for

postoperative ileus. Similar to the ICAM-1 antibody (anti-CD54 IgG2b clone YN1/1.7)24, 33,

ISIS-3082 reduces the IM-induced inflammatory cell influx and improves gastric emptying

in a dose-dependent manner, with a maximum effect at 1mg/kg, restoring delayed gastric

emptying. As the nonsense control oligonucleotide (ISIS 8997) in a dose of 1 as well 3mg/kg

did not have these effects, a sequence unspecific effect of the phosphorothioate backbone

can be excluded. Therefore, we conclude that the anti-inflammatory effect of ISIS-3082

observed results from a sequence-specific reduction in ICAM-1 mRNA translation and

protein expression. The latter is supported by the immunohistochemical staining showing a

reduction of ICAM-1 expression by ISIS-3082, but not by ISIS-8997 or saline.

In the pharmacological range tested, the anti-inflammatory effect of ISIS-3082 diminished

in higher doses (3 and 10 mg/kg). Bennett et al.27 observed a similar dosedependent effect

in a study evaluating ISIS-3082 in a DSS colitis model. The lack of effect of higher dosages

may be explained by the pro-inflammatory properties of the phosphorothioate backbones

in antisense oligonucleotides like ISIS-308227, 29, 34. However, ICAM-1 expression was not

reduced in the presence of the local muscle inflammation, making this possibility less

likely. A more plausible explanation might be the biphasic response of ribonuclease H

activity on phosphorothioate antisense oligonucleotide concentration. Low concentrations

of phosphorothioate oligonucleotides increase ribonuclease H activity, whereas high

concentrations have the opposite effect, leading to increased stability of the antisense-

bound mRNA35. The latter leads to decreased breakdown of ISIS-3082-bound (ICAM-1-

specific) mRNA by ribonuclease H, and a diminished effect on ICAM-1 protein synthesis.

At present, treatment of postoperative ileus consists of supportive measures such as nothing

by mouth, nasogastric suction, i.v. fluids, and the use of prokinetic and antiemetic drugs.

Unfortunately, this approach has been rather disappointing3, 36. Based on the current data,

pretreatment of patients with antibodies or antisense oligonucleotides targeted against

Page 61: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

60

ICAM-1 are possible new preventive strategies to shorten postoperative ileus. One of the

risks of using antibody treatment is the potential formation of neutralising antibodies18, 19.

Antisense oligonucleotides could represent an alternative to antibody treatment. The human

equivalent of ISIS-3082 (ISIS-2302) is currently being tested in a clinical trial as a putative

new treatment for inflammatory bowel disease. Based on the bell-shaped dose–response

curve, it should be emphasised that the therapeutic range is narrow, compromising its

clinical use. In addition, one should also consider that leukocyte recruitment to traumatised

tissues is needed for healing of the surgical wound. Both ISIS-3082 and ISIS-2302 have

been extensively tested in several, also surgeryinvolving, models, disorders and clinical

trials. None of theses studies reported impairment of wound healing or other postsurgical

complications32, 37, 38. In conclusion, ICAM-1 antisense pretreatment prevented postoperative

ileus in mice by reduction of manipulationinduced intestinal muscle inflammation. Our

data encourage further clinical evaluation of ICAM-1 antisense oligonucleotides as tool to

prevent postoperative ileus.

Page 62: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

3IC

AM

-1 A

ntise

nse

Olig

onuc

leot

ides

Pre

vent

Pos

tope

rativ

e Ile

us

61

Reference ListPrasad M, Matthews JB. Deflating postoperative ileus. Gastroenterology 1999;117:489-492.1. Livingston EH, Passaro EP, Jr. Postoperative ileus. Dig.Dis.Sci. 1990;35:121-132.2. Kehlet H, Holte K. Review of postoperative ileus. Am.J.Surg. 2001;182:3S-10S.3. Plourde V, Wong HC, Walsh JH, Raybould HE, Tache Y. CGRP antagonists and capsaicin on 4. celiac ganglia partly prevent postoperative gastric ileus. Peptides 1993;14:1225-1229.Barquist E, Bonaz B, Martinez V, Rivier J, Zinner MJ, Tache Y. Neuronal pathways involved in 5. abdominal surgery-induced gastric ileus in rats. Am.J.Physiol 1996;270:R888-R894.De Winter BY, Boeckxstaens GE, De Man JG, Moreels TG, Herman AG, Pelckmans PA. 6. Effects of mu- and kappa-opioid receptors on postoperative ileus in rats. Eur.J.Pharmacol. 1997;339:63-67.De Winter BY, Boeckxstaens GE, De Man JG, Moreels TG, Herman AG, Pelckmans PA. 7. Effect of adrenergic and nitrergic blockade on experimental ileus in rats. Br.J.Pharmacol. 1997;120:464-468.Boeckxstaens GE, Hirsch DP, Kodde A, Moojen TM, Blackshaw A, Tytgat GN, Blommaart PJ. 8. Activation of an adrenergic and vagally-mediated NANC pathway in surgery-induced fundic relaxation in the rat. Neurogastroenterol.Motil. 1999;11:467-474.Boeckxstaens GE, Hollmann M, Heisterkamp SH, Robberecht P, de Jonge WJ, van den Wi-9. jngaard RM, Tytgat GN, Blommaart PJ. Evidence for VIP(1)/PACAP receptors in the afferent pathway mediating surgery-induced fundic relaxation in the rat. Br.J.Pharmacol. 2000;131:705-710.Kalff JC, Buchholz BM, Eskandari MK, Hierholzer C, Schraut WH, Simmons RL, Bauer AJ. Bi-10. phasic response to gut manipulation and temporal correlation of cellular infiltrates and muscle dysfunction in rat. Surgery 1999;126:498-509.Kalff JC, Carlos TM, Schraut WH, Billiar TR, Simmons RL, Bauer AJ. Surgically induced leuko-11. cytic infiltrates within the rat intestinal muscularis mediate postoperative ileus. Gastroenterol-ogy 1999;117:378-387.de Jonge WJ, van den Wijngaard RM, The FO, ter Beek ML, Bennink RJ, Tytgat GN, Buijs 12. RM, Reitsma PH, van Deventer SJ, Boeckxstaens GE. Postoperative ileus is maintained by intestinal immune infiltrates that activate inhibitory neural pathways in mice. Gastroenterology 2003;125:1137-1147.Kreiss C, Birder LA, Kiss S, VanBibber MM, Bauer AJ. COX-2 dependent inflammation in-13. creases spinal Fos expression during rodent postoperative ileus. Gut 2003;52:527-534.Smith CW, Marlin SD, Rothlein R, Toman C, Anderson DC. Cooperative interactions of LFA-1 14. and Mac-1 with intercellular adhesion molecule-1 in facilitating adherence and transendothelial migration of human neutrophils in vitro. J.Clin.Invest 1989;83:2008-2017.Issekutz AC, Rowter D, Springer TA. Role of ICAM-1 and ICAM-2 and alternate CD11/CD18 15. ligands in neutrophil transendothelial migration. J.Leukoc.Biol. 1999;65:117-126.Dustin ML, Rothlein R, Bhan AK, Dinarello CA, Springer TA. Induction by IL 1 and interferon-16. gamma: tissue distribution, biochemistry, and function of a natural adherence molecule (ICAM-1). J.Immunol. 1986;137:245-254.Rothlein R, Dustin ML, Marlin SD, Springer TA. A human intercellular adhesion molecule 17. (ICAM-1) distinct from LFA-1. J.Immunol. 1986;137:1270-1274.Shawler DL, Bartholomew RM, Smith LM, Dillman RO. Human immune response to multiple 18. injections of murine monoclonal IgG. J.Immunol. 1985;135:1530-1535.LoBuglio AF, Wheeler RH, Trang J, Haynes A, Rogers K, Harvey EB, Sun L, Ghrayeb J, Khaz-19. aeli MB. Mouse/human chimeric monoclonal antibody in man: kinetics and immune response. Proc.Natl.Acad.Sci.U.S.A 1989;86:4220-4224.Crooke ST. Therapeutic applications of oligonucleotides. Annu.Rev.Pharmacol.Toxicol. 20. 1992;32:329-376.Stein CA, Cheng YC. Antisense oligonucleotides as therapeutic agents--is the bullet really 21. magical? Science 1993;261:1004-1012.

Page 63: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

62

Miner P, Wedel M, Bane B, Bradley J. An enema formulation of alicaforsen, an antisense 22. inhibitor of intercellular adhesion molecule-1, in the treatment of chronic, unremitting pouchitis. Aliment.Pharmacol.Ther. 2004;19:281-286.van Deventer SJ, Tami JA, Wedel MK. A randomised, controlled, double blind, escalating dose 23. study of alicaforsen enema in active ulcerative colitis. Gut 2004;53:1646-1651.Lub M, van Kooyk Y, Figdor CG. Competition between lymphocyte function-associated antigen 24. 1 (CD11a/CD18) and Mac-1 (CD11b/CD18) for binding to intercellular adhesion molecule-1 (CD54). J.Leukoc.Biol. 1996;59:648-655.Butler M, Stecker K, Bennett CF. Cellular distribution of phosphorothioate oligodeoxynucle-25. otides in normal rodent tissues. Lab Invest 1997;77:379-388.Crooke ST, Graham MJ, Zuckerman JE, Brooks D, Conklin BS, Cummins LL, Greig MJ, 26. Guinosso CJ, Kornbrust D, Manoharan M, Sasmor HM, Schleich T, Tivel KL, Griffey RH. Pharmacokinetic properties of several novel oligonucleotide analogs in mice. J.Pharmacol.Exp.Ther. 1996;277:923-937.Bennett CF, Kornbrust D, Henry S, Stecker K, Howard R, Cooper S, Dutson S, Hall W, Jacoby 27. HI. An ICAM-1 antisense oligonucleotide prevents and reverses dextran sulfate sodium-in-duced colitis in mice. J.Pharmacol.Exp.Ther. 1997;280:988-1000.Bennink RJ, de Jonge WJ, Symonds EL, van den Wijngaard RM, Spijkerboer AL, Benninga 28. MA, Boeckxstaens GE. Validation of gastric-emptying scintigraphy of solids and liquids in mice using dedicated animal pinhole scintigraphy. J.Nucl.Med. 2003;44:1099-1104.Pisetsky DS, Reich CF. Stimulation of murine lymphocyte proliferation by a phosphorothioate 29. oligonucleotide with antisense activity for herpes simplex virus. Life Sci. 1994;54:101-107.de Jonge WJ, The FO, van der CD, Bennink RJ, Reitsma PH, van Deventer SJ, van den 30. Wijngaard RM, Boeckxstaens GE. Mast cell degranulation during abdominal surgery initiates postoperative ileus in mice. Gastroenterology 2004;127:535-545.Kalff JC, Turler A, Schwarz NT, Schraut WH, Lee KK, Tweardy DJ, Billiar TR, Simmons RL, 31. Bauer AJ. Intra-abdominal activation of a local inflammatory response within the human mus-cularis externa during laparotomy. Ann.Surg. 2003;237:301-315.Stepkowski SM, Tu Y, Condon TP, Bennett CF. Blocking of heart allograft rejection by intercel-32. lular adhesion molecule-1 antisense oligonucleotides alone or in combination with other immu-nosuppressive modalities. J.Immunol. 1994;153:5336-5346.Pruijt JF, van Kooyk Y, Figdor CG, Lindley IJ, Willemze R, Fibbe WE. Anti-LFA-1 blocking an-33. tibodies prevent mobilization of hematopoietic progenitor cells induced by interleukin-8. Blood 1998;91:4099-4105.Zhao Q, Temsamani J, Iadarola PL, Jiang Z, Agrawal S. Effect of different chemically modified 34. oligodeoxynucleotides on immune stimulation. Biochem.Pharmacol. 1996;51:173-182.Gao WY, Han FS, Storm C, Egan W, Cheng YC. Phosphorothioate oligonucleotides are inhibi-35. tors of human DNA polymerases and RNase H: implications for antisense technology. Mol.Pharmacol. 1992;41:223-229.Luckey A, Livingston E, Tache Y. Mechanisms and treatment of postoperative ileus. Arch.Surg. 36. 2003;138:206-214.Kahan BD, Stepkowski S, Kilic M, Katz SM, Van Buren CT, Welsh MS, Tami JA, Shanahan 37. WR, Jr. Phase I and phase II safety and efficacy trial of intercellular adhesion molecule-1 anti-sense oligodeoxynucleotide (ISIS 2302) for the prevention of acute allograft rejection. Trans-plantation 2004;78:858-863.Chen W, Langer RM, Janczewska S, Furian L, Geary R, Qu X, Wang M, Verani R, Condon T, 38. Stecker K, Bennett CF, Stepkowski SM. Methoxyethyl-modified intercellular adhesion mole-cule-1 antisense phosphorothiateoligonucleotides inhibit allograft rejection, ischemic-reperfu-sion injury, and cyclosporine-induced nephrotoxicity. Transplantation 2005;79:401-408.

Page 64: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

3IC

AM

-1 A

ntise

nse

Olig

onuc

leot

ides

Pre

vent

Pos

tope

rativ

e Ile

us

63

Page 65: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

4

Page 66: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

4Cha

pter

4The vagal anti-inflammatory

pathway attenuates intestinal

macrophage activation and in-

flammation by nicotinic acetyl-

choline receptor mediated ac-

tivation of Jak-2/Stat-3.

Wouter J de Jonge, Esmerij P van der Zanden,

Frans O The, Maarten F Bijlsma,

David J van Westerloo, Roelof J Bennink,

Hans-Rudolf Berthoud, Satoshi Uematsu,

Shizuo Akira, Rene M van den Wijngaard

Guy E Boeckxstaens

Nature Immunology 2005; 6: 844-851

Page 67: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

66

AbstractAcetylcholine released by efferent vagus nerves inhibits macrophage activation. Here

we show that the anti-inflammatory action of nicotinic receptor activation in peritoneal

macrophages was associated with activation of the transcription factor STAT3. STAT3 was

phosphorylated by the tyrosine kinase Jak2 that was recruited to the α7 subunit of the

nicotinic acetylcholine receptor. The anti-inflammatory effect of nicotine required the ability

of phosphorylated STAT3 to bind and transactivate its DNA response elements. In a mouse

model of intestinal manipulation, stimulation of the vagus nerve ameliorated surgery-induced

inflammation and postoperative ileus by activating STAT3 in intestinal macrophages. We

conclude that the vagal anti-inflammatory pathway acts by α7 subunit−mediated Jak2-

STAT3 activation.

Page 68: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

4Va

gal A

nti-i

nflam

mat

ory

Path

way

Med

iate

d th

roug

h N

icot

inic

Jak

-2/S

tat-3

Act

ivat

ion

67

TBackgroundThe innate immune response has been increasingly recognized as being under substantial

neuronal control1. For example, acetylcholine or nicotine effectively attenuates the activation

of macrophages2. This so-called ‘cholinergic anti-inflammatory pathway’ is characterized

by a nicotine dose−dependent decrease in the production of proinflammatory mediators,

including high-mobility group box 1 proteins3, tumor necrosis factor (TNF), interleukin 1β (IL-

1β), IL-6 and IL-18 2, by macrophages stimulated with endotoxin. Consistently, stimulation of

the efferent vagus nerve dampens macrophage activation in rodent models of endotoxemia

and shock1, 2. Two nicotinic acetylcholine receptor (nAChR) subtypes are involved in the

nicotine-induced decrease in proinflammatory cytokine production by stimulated human

and mouse macrophages: the α7 homopentamer expressed by monocyte-derived

human and mouse macrophages4, and the α4β2 heteropentamer expressed by alveolar

macrophages5. Activation of the α7 homopentamer nAChR inhibits transactivational activity

of the transcription factor NF-κB p65 3. However, the subcellular mechanism explaining the

deactivating effect of acetylcholine on macrophages has remained unknown.

Here we evaluated the involvement of the transcription factor STAT3 in this process,

because STAT3 is a potential negative regulator of inflammatory responses6, 7. STAT3 and

the tyrosine kinase Jak2, which phosphorylates STAT3, are required for both IL-6 receptor

(IL-6R) and IL-10R signaling. IL-6 contributes to the progression of many inflammatory

diseases, whereas IL-10 is an anti-inflammatory cytokine that suppresses the activation of

macrophages. IL-6R signaling is inhibited by the Src homology 2 domain protein SOCS3,

whose expression is induced by STAT3 activation8, 9. SOCS3 binds to the glycoprotein 130

(gp130) subunit of the IL-6R, leading to inhibited activation of STAT3 by IL-6R ligands8, 9.

Consistent with that finding, in LPS-stimulated macrophages deficient in SOCS3, IL-6R

ligands induce a sustained STAT3 activation, which leads to the reduced production of

proinflammatory cytokines such as TNF10.

Here we demonstrate that nicotine exerts its anti-inflammatory effect on peritoneal

macrophages via Jak2 and STAT3 signaling in vitro and in vivo. In isolated peritoneal

macrophages, nicotine activated nAChRs, leading to phosphorylation of STAT3 via Jak2.

Page 69: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

68

Jak2 was recruited to the α7 subunit of the nAChR and was phosphorylated after nicotine

binding. We further studied the effect of cholinergic inhibition of macrophage activity in vivo

on the occurrence of post-surgical intestinal inflammation in a mouse model of postoperative

ileus11, 12. Postoperative ileus is characterized by general hypomotility of the gastrointestinal

tract and delayed gastric emptying13 and is a pathological condition commonly noted after

abdominal surgery with intestinal manipulation. This condition is the result of inflammation

of the intestinal muscularis due to activation of resident macrophages14, 15 that are triggered

by bowel manipulation12. We show here that perioperative stimulation of the vagus nerve

prevented manipulation-induced inflammation of the intestinal muscularis externa and

ameliorated postoperative ileus. The effectiveness of stimulation of the vagus nerve in

reducing intestinal inflammation depended on STAT3 activation in macrophages in the

intestinal muscularis. Hence, our data demonstrate the molecular pathway responsible for

cholinergic inhibition of macrophage activation and suggest that stimulation of the vagus

nerve or administration of cholinergic agents may be effective anti-inflammatory therapy for

the treatment of postoperative ileus and other inflammatory diseases.

Page 70: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

4Va

gal A

nti-i

nflam

mat

ory

Path

way

Med

iate

d th

roug

h N

icot

inic

Jak

-2/S

tat-3

Act

ivat

ion

69

Methods Reagents and antibodies.Nicotine, hexamethonium, α-bungarotoxin, methyllycaconitine citrate, d-tubocurarin,

dihydro-β-erythroidine, AG 490, cycloheximide, actinomycin-D and rat monoclonal

antibody to β2 nAChR subunit (anti-β2) were from Sigma-Aldrich. Polyclonal rabbit anti-

Jak2, anti−phosphorylated Jak2, anti-SOCS3 and anti-α7 were obtained from Abcam; goat

polyclonal anti-actin, rabbit polyclonal anti-STAT1 and rabbit polyclonal anti-STAT3 were

from Santa Cruz Biotechnology; and rabbit polyclonal anti−phosphorylated STAT1 and

anti-phosphorylated STAT3 were from Cell Signaling Technology. ELISA kits for IL-6, IL-10,

MIP-1α, MIP-2 and TNF were from R&D Systems.

Cell culture and transient transfection.Resident peritoneal macrophages were collected from BALB/c mice by flushing of the

peritoneal cavity with 5 ml of ice-cold Hank’s balanced salt solution containing 10 U/ml

of heparin. Peritoneal cells were plated at a density of 1 x 106 cells/cm2 in RPMI medium

supplemented with 10% FCS, and macrophages were left to adhere for 2 h in a humidified

atmosphere at 37 °C with 5% CO2. Cells were washed and the remaining macrophages were

left for 16−20 h. Subsequently, cells were preincubated with the appropriate concentration of

nicotine for 15 min, followed by challenge for 3 h with LPS (1−100 ng/ml). NAChR blockers

were added 30 min before nicotine, and no toxicity was noted after 4 h of incubation

with any blocker, as assessed by the trypan blue exclusion test. Cycloheximide (10 µg/

ml) and actinomycin-D (5 µg/ml) were added 5 min before nicotine. Cells were lysed for

immunoblots 30 min after exposure to nicotine and/or LPS. Peritoneal macrophages were

transfected with the Effectine reagent (Qiagen) according to the manufacturer's instructions.

A cytomegalovirus-driven Renilla luciferase reporter plasmid was cotransfected to allow

assessment of transfection efficiency. The pCAGGS-neo expression vectors encoding wild-

type hemagglutinin-tagged STAT3 or the dominant negative mutant hemagglutinin-tagged

STAT3D17 were provided by I. Touw (Erasmus University, Rotterdam, The Netherlands) and

T. Hirano (Osaka University, Osaka, Japan). In hemagglutinin-tagged STAT3D, glutamic

acids 434 and 435 were replaced by alanines17. After transfection, cells were selected for

16 h with neomycin (2.0 mg/ml; Sigma-Aldrich), were washed and were treated with nicotine

Page 71: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

70

and LPS 24 h after transfection. Transfection was verified by immunoblot with horseradish

peroxidase−tagged rabbit polyclonal anti-hemagglutinin (Abcam). For small interfering RNA

transfection, cells were transfected with a small interfering RNA oligonucleotide specific to

SOCS3 (ID 160220; Ambion) using RNAiFect (Qiagen) according to the manufacturer’s

instructions. A fluorescein isothiocyanate−labeled control random RNA oligonucleotide

(Ambion) was cotransfected to optimize transfection efficiency.

Immunoblots.Cells were scraped in 50 µl of ice-cold lysis buffer containing 150 mM NaCl, 0.5% Triton

X-100, 5 mM EDTA and 0.1% SDS. Samples were 'taken up' in 50 µl sample buffer (125

mM Tris-HCl, pH 6.8, 2% SDS, 10% β-mercaptoethanol, 10% glycerol and 0.5 mg/ml of

bromophenol blue), were separated by SDS-PAGE and were blotted onto polyvinyldifluoride

membranes (Millipore). Membranes were blocked in 0.1% Tween-20 in Tris-buffered saline

containing 5% nonfat dry milk and were incubated overnight with the appropriate antibodies

in 1% BSA and 0.1% Tween-20 in Tris-buffered saline. Horseradish peroxidase−conjungated

secondary antibodies were visualized with Lumilite plus (Boehringer-Mannheim).

Immunoprecipitation.Peritoneal macrophages at a density of 1 x 106 per cm2 were preincubated for 30 min

with 1 µM nicotine and 100 µM AG 490, were scraped in lysis buffer (20 mM Tris-HCl,

pH 7.6, 2.5 mM EDTA, 1 mM EGTA, 1% Triton X-100, 0.5% sodiumdeoxycholate, 10%

glycerol, 1 mM Na3VO4, 50 mM NaF, 1 µg/ml of aprotinin, 1 µg/ml of leupeptin and 1 mM

phenylmethyl sulfonyl fluoride), were sonicated for 10 s and were centrifuged at 4 °C for

20 min at 14,000g. Lysates preabsorbed to 20 µl protein A−protein G (Sigma-Aldrich)

were incubated overnight with the appropriate antibodies and were immunoprecipitated

with 40 µl protein A−protein G. Alternatively, the TrueBlot system (eBioscience)

was used for immunoprecipitation according to the manufacturer’s instructions.

Immunoprecipitates were recovered by centrifugation, were washed in ice-cold wash

buffer (0.1% Triton X-100 and 1 mM phenylmethyl sulfonyl fluoride in Tris-buffered saline)

and were ‘taken up’ in sample buffer (125 mM Tris-HCl, pH 6.8, 2% SDS, 10% glycerol

and 0.5 mg/ml of bromophenol blue), followed by immunoblot as described above.

Page 72: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

4Va

gal A

nti-i

nflam

mat

ory

Path

way

Med

iate

d th

roug

h N

icot

inic

Jak

-2/S

tat-3

Act

ivat

ion

71

Surgical procedures.Mice (female BALB/c) were used at 15−20 weeks of age. IL-6- and IL-10-deficient mice and

their respective C57BL/6 wild-type counterparts were obtained from Jackson Laboratories.

LysM-Cre Stat3fl/fl and Stat3fl/fl mice were maintained at Osaka University (Osaka, Japan).

Abdominal surgery with intestinal manipulation was done as described elsewhere11. Mice

(n = 10−12) were assigned to the following four groups: control surgery of laparotomy

only; laparotomy followed by intestinal manipulation combined with sham preparation of

the cervical area; laparotomy in combination with electrical stimulation of the vagus nerve;

or intestinal manipulation in combination with electrical stimulation of the vagus nerve.

Intestinal manipulation consisted of 5 min of manipulation of the distal duodenum to the

cecum with sterile moist cotton applicators. At 3 or 24 h after surgery, mice were killed

by cervical dislocation. The small intestine was removed, flushed and fixed in ice-cold

100% ethanol for the preparation of whole mounts. Small intestinal muscularis strips were

prepared by pinning of freshly isolated intestinal segments in ice-cold PBS and removal

of mucosa facing upward. Muscle strips were ‘snap-frozen’ in liquid nitrogen and were

stored at − 80 °C until analysis. All animal experiments were in compliance with guidelines

set by the Animal Ethics Committee of the University of Amsterdam (Amsterdam, The

Netherlands).

Electric stimulation of the vagus nerve.Stimulation of the vagus nerve was essentially done as described2. The left cervical nerve

was prepared free from the carotid artery and was ligated with 6-0 silk suture. The distal

part of the ligated nerve trunk was placed in a bipolar platinum electrode unit. In some

experiments, the vagus nerve was transected and the distal part was stimulated. Voltage

stimuli (5 Hz for 2 ms at 1 or 5 V) were applied for 5 min before and for 15 min after the

intestinal manipulation protocol described above. For sham stimulation of the vagus nerve,

in control mice the cervical skin was opened and was covered by moist gaze for 20 min.

Local blockade of nicotinic receptors in the ileum was done as follows: in anesthetized

mice (n = 7), a midline laparotomy incision was made and 6 cm of ileum proximal to the

cecum was carefully externalized and placed in a sterile preheated tube. The segment

was incubated for 20 min with a preheated (37 °C) solution of hexamethonium (100 µM

Page 73: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

72

in 0.9% NaCl) or vehicle. The temperature of the intestinal tissue was monitored with a

thermal probe. Leakage of hexamethonium solution into the peritoneal cavity was strictly

avoided. After incubation, the hexamethonium solution was removed and the ileal segment

was washed three times with 0.9% NaCl and was included in the manipulation protocol.

Measurement of gastric emptying.Gastric emptying of a semiliquid, noncaloric ‘test meal’ (0.5% methylcellulose) containing

10 MBq 99mTc was assessed by scintigraphic imaging as described37.

Quantification of leukocyte accumulation at the intestinal muscularis.Myeloperoxidase activity in ileal muscularis tissue was assayed as a measure of leukocyte

infiltration as described11, 23. Whole mounts of ethanol-fixed ileal muscularis were prepared

and stained for myeloperoxidase activity as described11, 23.

RT-PCR. Total RNA from tissue was isolated with Trizol (Invitrogen), treated with DNase and re-

verse-transcribed. The resulting cDNA (0.5 ng) was subjected to 40 cycles of Light Cycler

PCR (FastStart DNA Master SYBR Green; Roche). The primers used were as follows:

TNF antisense, 5’-AAAGCATGAT CCGCGACGT-3’, and sense, 5’-TGCCACAAGCAG-

GAATGAGAA-3’; MIP-2 antisense, 5’-AGTGAACTGCGCTGTCAATGC-3’, and sense,

5’-GCAAACTTTTTGACCGCCCT-3’; SOCS3 antisense, 5’-ACCTTTCTTATCCGCGA-

CAG-3’, and sense, 5’-TGCACCAGCTTGAGTACACAG-3’; and glyceraldehyde phospho-

dehydrogenase (GAPDH) antisense, 5’- ATGTGTCCGTCGTGGATCTGA-3’, and sense,

5’-ATGCCTGCTTCACCACCTTCT-3’. PCR products were quantified with a linear regres-

sion method using the Log(fluorescence) per cycle number38 and data are expressed as

the percentage of GAPDH transcripts for each sample. For qualification, the resulting PCR

products were separated by 2.5% agarose gel electrophoresis and analyzed by ethidium

bromide staining.

Immunohistochemistry.For double-labeling of macrophages and cholinergic fibers, Sprague-Dawley rats

(300−350 g; Harlan Industries) were anesthetized with pentobarbital sodium (90 mg/

kg intraperitoneally) and were perfused transcardially with heparinized saline (20 U/

Page 74: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

4Va

gal A

nti-i

nflam

mat

ory

Path

way

Med

iate

d th

roug

h N

icot

inic

Jak

-2/S

tat-3

Act

ivat

ion

73

ml) followed by ice-cold 4% phosphate-buffered paraformaldehyde, pH 7.4. Gastric and

intestinal tissue were extracted and were postfixed for a minimum of 2 h in the same

fixative. Tissue was cryoprotected overnight in 18% sucrose and 0.05% sodium azide in

0.01 M PBS. Flat sections 20 µm in thickness and cross-sections 25 µm in thickness of

the corpus and mid ileum were cut on a cryostat and were processed in PBS. Sections

were pretreated with 0.5% sodium borohydride in PBS and were subsequently blocked in

donkey normal serum. Monoclonal mouse anti−rat CD163 (ED2; Serotec) and polyclonal

goat anti−vesicular acetylcholine transporter (Chemicon) were diluted in 0.1% gelatin and

0.05% sodium azide in PBS with 0.5% Triton X-100 and were incubated for 20 h at 20 °C

or for 48 h at 4 °C. Secondary antibodies used were indocarbocyanine-conjugated donkey

anti-mouse (Jackson ImmunoResearch) for ED2 and carbocyanine-conjugated donkey

anti-goat (Jackson Immuno-Research) for vesicular acetylcholine transporter in 0.05%

sodium azide in PBS with 0.5% Triton X-100. Sections were mounted in 100% glycerol with

the addition of 5% N-propyl gallate as an antifade agent.

In vivo labeling of mouse phagocytes was achieved by intraperitoneal injection of 20 µg

Alexa 546−labeled dextran particles (molecular weight, 10,000; Molecular Probes) 24 h

before surgery. At 1 h after surgery, anesthetized mice were perfused with 10 ml of ice-cold

0.9% NaCl containing 1 mM Na3VO4, followed by 20 ml of ice-cold 4% formaldehyde solution,

pH 7.4. Intestinal tissue was isolated, fixed overnight in 4% formaldehyde, dehydrated

and embedded in paraffin. Sections 6 µm in thickness were cut and were immunostained

with polyclonal rabbit anti−phosphorylated STAT3 (Cell Signaling Technologies) and biotin-

labeled anti-rabbit according to the manufacturer’s instructions. Biotin was visualized with

3-amino-9-ethyl carbazole (Sigma) as a chromogen, followed by counterstaining with

hematoxylin. Alternatively, Alexa 488−streptavidin (Molecular Probes) with 4,6-diamidino-

2-phenylindole nuclear counterstain was used for analysis by confocal microscopy.

Statistics.Statistical analysis of the results was performed by variance followed by Dunnett’s post-

hoc test or nonparametric Mann-Whitney U tests with SPSS. A probability value (P) of less

than 0.05 was considered significant.

Page 75: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

74

Results Nicotine activates STAT3 in macrophagesTo study the cellular response of macrophages to nicotinic receptor activation, we isolated

peritoneal macrophages from mice and investigated the effect of nicotine on LPS-induced

cytokine production. Nicotine reduced the LPS-induced release of TNF, MIP-2 and IL-6 but

not IL-10 in a dose-dependent way (Fig. 1a), consistent with published reports on the anti-

inflammatory effect of nicotine on human and mouse monocyte-derived macrophages2, 4, 5.

Given the crucial function of STAT3 in anti-inflammatory responses6, 7, we hypothesized

that activation of STAT3 and its gp130-binding regulatory protein SOCS3 may be involved

in the anti-inflammatory effect of nicotine. Consistent with that hypothesis, we found that

nicotine treatment activated STAT3 as well as SOCS3 in resting and LPS-stimulated

primary peritoneal macrophages in a dose- and time-dependent way (Fig. 1b,c). Nicotine

activated STAT3 directly, as phosphorylation of STAT3 was not affected by the protein

synthesis inhibitors actinomycin D and cycloheximide (Fig. 1d). In contrast, interferon-γ-

induced STAT1 activation was not effected by nicotine (Fig. 1e). Thus, nicotine reduced

the production of proinflammatory cytokines and activated STAT3 as well as SOCS3 in

stimulated macrophages.

A

0.4

0.6

0.8

Nicotine (nM)

Cyt

okin

e (n

g/m

l)

IL-10

0.1

0.2

0.3

0.4

1

2

3

IL-6TNF

2

4

6MIP-2

0 100 10 102 103 0 100 10 102 103 0 100 10 102 103 0 100 10 102 103

Page 76: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

4Va

gal A

nti-i

nflam

mat

ory

Path

way

Med

iate

d th

roug

h N

icot

inic

Jak

-2/S

tat-3

Act

ivat

ion

75

No LPS LPS (1 ng/ml)

B

PY-STAT3

Actin

0 101 102 103

STAT3

nicotine (nM) 0 101 102 103

C

STAT3

PY-STAT3

STAT3

Time (min)

PY-STAT3

Time (min)Nicotine ( 100nM)

Vehicle0 10 20 30 40 60

0 10 20 30 40 60

SOCS3

g

(

S

Time (miot e 00 M

hi

nicotine(100nM)

D

STAT3

– +

Vehicle

+ – +

Act-D CHX

SOCS3

PY-STAT3

Nicotine (nM)

Actin

PY-STAT1

STAT1

0 10 102 103

Vehicle IFN- 100 ng/ml0 10 102 103

E

Actin

Figure 1. Nicotine attenuates peritoneal macrophage activation and induces phosphorylation of STAT3 and SOCS3 expression.(a) ELISA of TNF, MIP-2, IL-6 and IL-10 in the supernatants of peritoneal macrophages stimulated with 100 ng/ml of LPS in vitro in the presence of nicotine (dose, horizontal axes). Data represent mean ± s.e.m. of four independent experiments in triplicate. (b) Immunoblots for phosphotyrosine-STAT3 (PY-STAT3), STAT3 and SOCS3 in cell lysates of peritoneal macrophages stimulated with 1 ng/ml of LPS (right) or no LPS (left) in the presence of nicotine (concentration, above lanes). Blot is representative of five independent experiments. (c) Immunoblot of phosphorylated STAT3 (PY-STAT3) and STAT3 in cell lysates of peritoneal macrophages stimulated with 100 nM nicotine (time, above lanes). Blot is one representative of three independent experiments. (d) Immunoblot of phosphorylated STAT3 (PY-STAT3), STAT3 and SOCS3 in cell lysates of peritoneal macrophages pretreated with vehicle, actinomycin-D (Act-D) or cycloheximide (CHX) and incubated with saline (-) or 100 nM nicotine (+). Blot is representative of three independent experiments. (e) Immunoblot of phosphorylated STAT1 (PY-STAT1) and STAT1 in peritoneal macrophages incubated with nicotine (concentration, above lanes) and stimulated with 100 ng/ml of interferon-γ (IFN-γ). Actin, loading control.

Page 77: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

76

Deactivation by nicotine requires STAT3 transactivationWe next sought to determine whether the anti-inflammatory effect of nicotine depended on

nuclear transactivation of phosphorylated STAT3. We overexpressed a dominant negative

form of STAT3 (STAT3D) in primary peritoneal macrophages. Dimerized STAT3D is altered

in its ability to bind DNA response elements and induce transcription of target genes16,

17. Nicotine failed to reduce LPS-induced TNF release in LPS-stimulated macrophages

transfected with STAT3D but not those transfected with the STAT3 wild-type construct (Fig.

2a). Thus, the nicotine-induced inhibition of TNF release is dependent on STAT3 DNA

transactivation.

To evaluate whether SOCS3 expression is crucial to the nicotinic anti-inflammatory effect,

we abrogated SOCS3 expression in peritoneal macrophages with SOCS3-specific small

interfering RNA (Fig. 2b). SOCS3 expression was substantially decreased in response to

nicotine (less than 10% of that expressed in control transfected cells), whereas STAT3

activation was not affected (transfection efficiency was more than 90%; Fig. 2b). In

macrophages with reduced SOCS3, however, nicotine was still able to decrease endotoxin-

induced production of IL-6 (data not shown) and TNF in a concentration-dependent way,

although the reduction was less pronounced than that in control transfected cells (Fig.

2c). Thus, blockade of STAT3 transactivation counteracted the anti-inflammatory effects

of nicotine, whereas blockade of SOCS3 expression did not. These results indicate that

SOCS3 expression is not strictly required for the reduction in macrophage TNF release by

nicotine.

Figure 2. Inhibition of macrophage activation by nicotine requires transactivation of STAT3 but not SOCS3 expression.(a) TNF in the supernatants of peritoneal macrophages transiently transfected with dominant nega-tive STAT3D, wild-type STAT3 (STAT3 WT)17 or empty vector (Vector), then incubated with nicotine and stimulated with 10 ng/ml of endotoxin. Values are expressed as the percent of TNF released without the addition of nicotine for each group. Data are mean s.e.m. of three independent experi-ments done in duplicate. *, P < 0.05 (one-way analysis of variance followed by Dunnett’s multiple comparison test). (b) Immunoblot for phosphorylated STAT3 (PY-STAT3), STAT3 and SOCS3 in peritoneal macrophages transiently transfected with control oligonucleotide or SOCS3-specific small interfering RNA (siRNA), then incubated with 100 nM nicotine. Blot is representative of three inde-pendent experiments. (c) TNF in the culture supernatants of peritoneal macrophages transfected with control oligonucleotide or SOCS3 siRNA, then preincubated with nicotine and stimulated with 10 ng/ml of LPS. Data are presented as percentage of TNF produced without addition of nicotine for each treatment group and are the mean ± s.e.m. of three independent experiments done in duplicate.

Page 78: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

4Va

gal A

nti-i

nflam

mat

ory

Path

way

Med

iate

d th

roug

h N

icot

inic

Jak

-2/S

tat-3

Act

ivat

ion

77

A

*

Nicotine (nM)

Perc

enta

ge d

ecre

ase

in T

NF

0 10-1 100 101 102 103

VectorSTAT3WTSTAT3D

siRNA

Con

trol

SOC

S3

Con

trol

SOCS3

PY-STAT3Nicotine (nM) 0 102 102

STAT3

B

0

100

20

60

80

40

Perc

enta

ge d

ecre

ase

in T

NF

Nicotine (nM)0 100 101 102

ControlSOCS3

C

0

100

20

60

80

40

Page 79: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

78

STAT3 phosphorylation depends on α7 nAChR activationTo determine whether STAT3 activation by nicotine was mediated by nAChR, we

pretreated cells with nAChR antagonists. The nonselective antagonists hexamethonium

and d-tubocurarine prevented the STAT3 phosphorylation induced by nicotine (Fig. 3a).

In addition, the α7 nAChR−selective antagonists α-bungarotoxin and methyllycaconitine

blocked the nicotine-induced STAT3 activation (Fig. 3a). A prominent function for the α7

receptor in nicotine-induced deactivation of macrophages corroborates published reports

on human and mouse monocyte-derived macrophage cultures3, 4. The selective non-α7

nAChRv antagonist dihydro-β-erythroidine did not affect nicotine-induced STAT3 activation

(data not shown).

Blocking nAChR also counteracted the attenuation of proinflammatory mediator release by

nicotine in activated macrophages. Hexamethonium, d-tubocurarine and methyllycaconitine

prevented the reduction in endotoxin-induced release of IL-6 (Fig. 3b) and MIP-2 (data not

shown) by nicotine in a dose-dependent way. Hexamethonium (effective dose leading to

50% inhibition (ED50), 6.46 ± 2.90 nM) was more potent than methyllycaconitine (ED50, 24.0

± 3.4 nM) and was far more potent than d-tubocurarine (ED50, 0.80 ± 0.23 µM) in attenuating

the inhibition of IL-6 release (Fig. 3b). The high ED50 for d-tubocurarine is probably due to

its low affinity for α7 nAChRs18 and is in line with its modest inhibitory effect on STAT3

activation by nicotine (Fig. 3a). In addition to methyllycaconitine, α-bungarotoxin abolished

IL-6 reduction by nicotine. However, exposure of the cells to α-bungarotoxin decreased

IL-6 production in the presence and absence of nicotine, which compromised adequate

determination of its ED50 (data not shown). Thus, STAT3 activation is dependent on the

activation of nAChRs by nicotine, most likely exclusive through activation of the α7 nAChR

subunit

fig 3

Actin

PY-STAT3

STAT3

Bgt(1 mg/ml)

d-TC (1 µM)

MLA(1 µM)

Hexa (1 µM)

A

0 101 102 103Nicotine (nM) 0 101 102 103 0 101 102 103 0 101 102 103 0 101 102 103

Page 80: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

4Va

gal A

nti-i

nflam

mat

ory

Path

way

Med

iate

d th

roug

h N

icot

inic

Jak

-2/S

tat-3

Act

ivat

ion

79

The macrophage α7 nAChR recruits Jak2STAT3 phosphorylation normally requires activity of the cytoplasmic tyrosine kinase Jak2 8.

Therefore, we investigated whether STAT3 phosphorylation depended on Jak2 activity and

whether nAChRs expressed on macrophages recruit Jak2. Phosphorylation of STAT3 after

nicotine treatment of peritoneal macrophages was effectively blocked by AG 490, a selective

inhibitor of Jak2 phosphorylation19, 20 (Fig. 4a). In agreement with that finding, nicotine failed

to reduce IL-6 release by LPS-stimulated peritoneal macrophages treated with AG 490

(data not shown). Binding studies have distinguished two main categories of nAChRs

based on their affinity for either α-bungarotoxin (α7-containing homopentamers) or nicotine

(α4β2 pentamers)18. Because our blocking studies suggested involvement of the α7 nAChR

subtype, we analyzed putative associations of α7 with Jak220 by immunoprecipitation (Fig.

4b). The α7 (56-kilodalton)21 receptor was expressed in primary peritoneal macrophage

lysates. Immunoprecipitation of Jak2 from peritoneal macrophage cell lysates showed a

weak association of Jak2 with the α7 receptor after culture in the absence of nicotine. To

investigate whether Jak2 is recruited to the nAChR and is phosphorylated after binding of

its ligand, we preincubated cells with nicotine. Nicotine exposure increased the amount of

α7 nAChR detected in Jak2 and phosphorylated Jak2 immunoprecipitates (Fig. 4b).

e Jon e

B

0

40

100

0 100 101 102 103 104

nAchR blocker (nM)

IL-6

inhi

bitio

n (%

)

6080

20

120

Figure 3. STAT3 phosphorylation by nicotine is prevented by α7-selective nAChR antagonists.(a,b) Peritoneal macrophages were pretreated with the nAChR blockers d-tubocurarin (d-TC), α-bungarotoxin (αBgt), hexamethonium (Hexa) or α-methyllycaconitine (MLA) and were incubated with nicotine (concentration, above lanes). Lysates were collected for immunoblot of phosphorylated STAT3 (PY-STAT3), STAT3 and actin (a) and IL-6 was measured in superna-tants (b). (a) Blots are representative of three independent experiments. (b) Filled squares, hexamethonium; open squares, methyllycaconitine; open circles, d-tubocurarine). Data are pre-sented as the percentage of inhibition of IL-6 release measured without the addition of an nAChR blocker and rep-resent mean values ± s.e.m. of three independent experiments done in trip-licate.

Page 81: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

80

+AG 490 (10 M)vehicle (1%EtOH) +AG 490 (100 M)

PY-STAT3

STAT3

A

0 101 102 103Nicotine (nM) 0 101 102 103 0 101 102 103

B

AG490 (100 M)IB:

7

PY-Jak2

IP: 7

Lysa

te Nicotine (1 M)++

+–– –

Igh

Jak2

7

Igh

IP: Jak2

1 2 3 4 5

IP: PY-Jak2

7

No

lysa

te

Igh

PY-Jak2

Jak2

Figure 4. Nicotine-induced STAT3 phosphorylation occurs through activation of Jak2 that is recruited to the α7 nAChR subunit. (a) Immunoblot of phospho-rylated STAT3 (PY-STAT3) and STAT3 in peritoneal macrophages incubated with AG 490 (concentrations, above blots). Blot is representative of three indepen-dent experiments. (b) Immunoblots of peritoneal macrophages treated with 1 M nicotine (lanes 4 and 5) or with 1 μM nicotine plus 100 μM AG 490 (lane 5). Cell lysates were immunoprecipitated (IP) with anti-α7 (top), anti-Jak2 (middle) or anti−phosphorylated Jak2 (PY-Jak2; bottom), followed by immunoblot (IB; antibodies, left margin). Lane 2, coprecipitate in the absence of lysate (negative control). IgH, immuno-globulin heavy chain. Blots are representative of four independent experiments.

Page 82: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

4Va

gal A

nti-i

nflam

mat

ory

Path

way

Med

iate

d th

roug

h N

icot

inic

Jak

-2/S

tat-3

Act

ivat

ion

81

To further demonstrate that Jak2 is phosphorylated after nAChR activation, we pretreated

cells with the Jak2 phosphorylation blocker AG 490 before adding nicotine. Cells treated

with AG 490 had reduced phosphorylated Jak2 in α7 immunoprecipitates, whereas Jak2

recruitment to the α7 receptor was not affected (Fig. 4b). The latter finding demonstrates

that Jak2 is recruited and phosphorylated after nicotine binding.

Stimulation of the vagus nerve ameliorates inflammationWe next evaluated whether activation of nAChR on macrophages would attenuate intestinal

inflammation in vivo. We assessed the effect of stimulation of the vagus nerve on the

inflammation that follows intestinal manipulation in our mouse model11, because this immune

response is associated with the activation of macrophages12, 22. We electrically stimulated

the left cervical vagus nerve during intestinal manipulation surgery and investigated the

effects on muscular inflammation and gastric emptying 24 h later (Fig. 5). Consistent with

published findings11, 23, intestinal manipulation of mice resulted in a delayed gastric emptying

compared with that of mice that underwent only laparotomy, indicative of the development

of postoperative ileus11 (gastric retention after 60 min, 14.5% ± 2.7% for laparotomy and

43.0 ± 6.7% for intestinal manipulation). However, stimulation of the vagus nerve prevented

the intestinal manipulation−induced gastroparesis 24 h after surgery (gastric retention,

25.2% ± 3.2%; Fig. 5). Notably, stimulation of the vagus nerve in itself may alter gastric

emptying during the vagus stimulation protocol24. However, we found that stimulation of

the vagus nerve did not affect basal gastric emptying 24 h after surgery (gastric retention,

15.7% ± 3.6%; Fig. 5). The last finding demonstrates that normalization of gastric emptying

after stimulation of the vagus nerve was not a direct effect on gastric motility but resulted

from reduced inflammation of the manipulated bowel segment11.

We next analyzed muscularis tissue for granulocytic infiltrates by measuring myeloperoxidase

activity in muscularis tissue homogenates and quantifying cellular infiltrates (Fig. 6). The

intestinal manipulation−induced inflammation of the muscularis externa in mice that received

stimulation of the vagus nerve was reduced in a voltage-dependent way compared with

that of mice that received intestinal manipulation plus sham stimulation. Prior vagotomy

of the proximal end of the stimulated vagus nerve did not affect these results (data not

shown), indicating that the anti-inflammatory effect of stimulation of the vagus nerve was

not dependent on the activation of central nuclei, which confirms published reports2.

Page 83: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

82

We next incubated intestinal segments with the nicotinic receptor blocker hexamethonium

before intestinal manipulation combined with stimulation of the vagus nerve. In intestinal

segments treated with hexamethonium, stimulation of the vagus nerve failed to prevent

inflammation, in contrast to incubation with vehicle (Fig. 6), demonstrating that the

anti-inflammatory effect of vagus stimulation acted through local activation of nicotinic

receptors.

20

40

60

80

100

0 20 40 60 800

Rel

ativ

e ga

stric

con

tent

(%)

*

Time after oral gavage (min)

Figure 5. Perioperative electrical stimulation of the left cervical va-gus nerve prevents gastroparesis 24 h after surgery with intestinal manipulation in mice.Gastric emptying curves of a semi-liquid ‘test meal’ are for mice that underwent surgery with intesti-nal manipulation (filled circles), control laparotomy surgery (gray triangles), stimulation of the va-gus nerve plus control laparotomy surgery (gray diamonds) or stimu-lation of the vagus nerve plus sur-gery with intestinal manipulation (filled squares). Values are means ± s.e.m.; n = 8−10. *, P < 0.05 for gastric retention at 60 min, surgery with intestinal manipulation versus surgery with intestinal manipulation plus stimulation of the vagus nerve (Mann-Whitney U test).

Page 84: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

4Va

gal A

nti-i

nflam

mat

ory

Path

way

Med

iate

d th

roug

h N

icot

inic

Jak

-2/S

tat-3

Act

ivat

ion

83

Figure 6. (see fullcolor chapter 11) Vagal nerve stimulation reduces recruitment of inflammatory infiltrates to the intestinal muscularis by activating peripheral nicotinic acetylcholine receptors. MPO activity measured in intestinal muscularis tissue homogenates isolated 24 h after surgery with IM. VNS with 5V, but not 1V, -stimulus prevents the increased muscularis MPO activity elicited by IM. Asterisks indicate significant differences in MPO activity in intestinal muscularis tissue from L control and IM VNS5V determined by one-way ANOVA followed by Dunnett’s multiple comparison test. Data represent mean ± SEM of 10-15 mice (a). MPO-activity containing cells were stained in whole mount preparations of intestinal muscularis (b and c) prepared 24 hrs post-operatively . Mice underwent IM with sham VNS (IM Sham), or IM combined with VNS using 1, or 5 V pulses (IM VNS1V, and IM VNS5V) (b). Mice were pretreated with hexamethonium (100 M; Hexa) or vehicle and underwent Laparotomy (L) with VNS (L VNS5V) or IM with VNS5V (e). MPO-positive cells were counted in five consecutieve microscopic fields of whole mount preparations of the indicated groups. Asterisks in-dicate significant differences (P<0.05) from (left graph) L control and (right graph) IM VNS5Vgroups using one-way ANOVA followed by Dunnett’s multiple comparison test. Data represent mean ± SEM of 5-8 mice.

Page 85: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

84

Stimulation of the vagus nerve activates STAT3 in vivoTo further investigate whether macrophages mediated the anti-inflammatory effect of

stimulation of the vagus nerve, we analyzed the expression of transcripts of macrophage-

derived inflammatory mediators in muscularis tissue 3 h after surgery. Stimulation of the

vagus nerve reduced the expression of Cxcl2 mRNA (Fig. 7a,b) and Ccl3 mRNA (data

not shown) but did not notably alter the expression of Tnf transcripts in muscularis tissue,

confirming earlier reports2, 4. However, when we analyzed peritoneal lavage fluid for the

presence of macrophage inflammatory mediators 3 h after intestinal manipulation, we found

that stimulation of the vagus nerve significantly reduced the secretion of TNF, IL-6, MIP-2

(Fig. 7c) and MIP-1α (data not shown) in the peritoneal cavity. This reduction was not due

to enhanced expression of IL-10, as stimulation of the vagus nerve was similarly potent in

reducing intestinal manipulation−induced inflammation in IL-10-deficient mice (Fig. 7d).

Moreover, the peritoneal IL-10 in wild-type mice did not reach the limit of detection (31 pg/

ml) at 1, 3 or 6 h after intestinal manipulation (data not shown). Expression of Socs3 (Fig.

7a) but not Socs1 (data not shown) was increased in muscularis tissue after stimulation of

the vagus nerve even in mice that underwent this stimulation without manipulation of the

bowel.

Tnf

Gapdh

Lsham

LVNS

IMsham

IMVNS

NoRT

-123

-107

-121

-132

Tnf

0

2

4

Lsham

LVNS

IMsham

IMVNS

Rel

ativ

e m

RN

A ex

pres

sion

Cxcl2

0

10

20

Lsham

LVNS

IMsham

IMVNS

Socs3

0

5

10

Lsham

LVNS

IMsham

IMVNS

Cxcl2

Socs3

B

A

* *

*

*

(bp)

Page 86: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

4Va

gal A

nti-i

nflam

mat

ory

Path

way

Med

iate

d th

roug

h N

icot

inic

Jak

-2/S

tat-3

Act

ivat

ion

85

Given the short half-life of acetylcholine, cholinergic regulation of macrophage activation

most likely requires that cholinergic nerves be in close proximity to intestinal macrophages.

To investigate this, we immunohistochemically double-labeled vesicular acetylcholine

transporter−positive vagal efferent fibers and macrophages in rat intestinal musclaris tissue.

f

2

4R

le

N

xe

io

Cxcl2

1

Socs3C

ytok

ine

(pg/

ml) MIP-2

0

800

1,600 *TNF

0

20

40 *

IL-6

(pg/

ml)

C

t

*

IL-6

0

400

800*

NDL

shamL

VNSIM

shamIM

VNSL

shamL

VNSIM

shamIM

VNSL

shamL

VNSIM

shamIM

VNS

Il10 –/–

0

300

600

Lsham

LVNS

IMsham

IMVNS

*

*D Il10 –/–

0

300

800

Lsham

LVNS

IMsham

IMVNS

*

*

Cel

l den

sity

(cel

ls/m

m2 )

ND

Figure 7. Vagal stimulation reduces intestinal manipulation-induced proinflammatory mediator ex-pression and release in vivo, independent of IL-10 production.(a,b) Real-time PCR for macrophage proinflammatory mediators (a, left margin; b, above graphs) of RNA isolated from intestinal muscularis strips prepared 3 h after the following procedures: control laparotomy surgery plus sham stimulation of the vagus nerve (L sham); control laparotomy surgery plus stimulation of the vagus nerve with 5-V pulses (L VNS); surgery with intestinal manipulation plus sham stimulation of the vagus nerve (IM sham); or surgery with intestinal manipulation plus stimu-lation of the vagus nerve with 5-V pulses (IM VNS). (a) No RT, no reverse transcriptase added to reaction (to control for nonspecific amplification); bp, base pairs. (b) Quantification38 of data and nor-malization of results to the expression of GAPDH. (c) Release of macrophage proinflammatory me-diators into peritoneal lavage fluid obtained 3 h after treatment of mice with the procedures described in a,b. (d) IL-6 in peritoneal cavities (left) and myeloperoxidase-positive cells intestinal muscularis tissues (right) of IL-10-deficient mice (open bars) and their wild-type counterparts (filled bars) after treatment with the procedures described in a,b. Right, myeloperoxidase-positive cells were quantified in whole-mount preparations of intestinal muscularis tissue isolated 24 h after the procedures. *, P < 0.05, compared with the respective control laparotomy surgery group (one-way ANOVA followed by Dunnett’s multiple comparison test (b,c) or Mann Whitney U test (d)). Data represent mean ± s.e.m. of five to eight mice. ND, not detectable.

Page 87: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

86

Macrophages were in close proximity to nerve terminals in the myenteric plexus in the

ileum (Fig. 8a) and circular muscle of gastric corpus (data not shown). Hence, acetylcholine

released from efferent nerve terminals could easily reach macrophages in the nanomolar

concentration range.

To verify that the enhanced SOCS3 expression reflected increased STAT3 activation in vivo,

we immunohistochemically analyzed intestinal tissues for the presence of phosphorylated

STAT3 in mice that underwent control laparotomy surgery, intestinal manipulation alone

or intestinal manipulation plus stimulation of the vagus nerve (Fig. 8b,c). We found

phosphorylated STAT3−positive nuclei in mice that underwent control laparotomy (Fig. 8b).

Intestinal manipulation resulted in the appearance of phosphorylated STAT3−positive cells

adhering to the serosal site of the bowel wall, most probably granulocytes and monocytes

recruited to the peritoneal compartment as a result of tissue trauma inflicted by the intestinal

manipulation procedure. However, when stimulation of the vagus nerve was applied, we

noted phosphorylated STAT3−positive nuclei in cells between longitudinal and circular

muscle layers surrounding the myenteric plexus. To identify the cellular source of the

phosphorylated STAT3−positive nuclei, we labeled tissue phagocytes in vivo by pretreating

mice with Alexa 546−labeled dextran particles (molecular weight, 10,000). This procedure

labels F4/80 antigen−positive macrophages populating the intestinal muscularis25. Most

of phosphorylated STAT3−positive nuclei in intestinal tissue of mice that had undergone

stimulation of the vagus nerve localized together with cells that had taken up Alexa

546−labeled dextran particles, indicating that these phosphorylated STAT3−positive nuclei

represented macrophages (Fig. 8c). These observations corroborate our in vitro findings

on the function of STAT3 in the cholinergic inhibition of tissue macrophages and are in

line with our proposed function of the network of resident intestinal macrophages26 as the

inflammatory cells targeted by stimulation of the vagus nerve. To further demonstrate that

the cholinergic anti-inflammatory pathway critically depends on STAT3 activation in vivo, we

studied the inflammatory response to intestinal manipulation in mice specifically deficient

in STAT3 in their myeloid cell lineage (called ‘LysM-Stat3fl/-’ mice here). LysM-Stat3fl/- mice

lack STAT3 in their macrophages and granulocytes6. In Stat3fl/+ control mice as well as in

LysM-Stat3fl/- mice, intestinal manipulation led to increased peritoneal IL-6 (Fig. 9a) as well

as massive inflammatory infiltrates in the manipulated muscularis tissue (Fig. 9b). Notably,

however, stimulation of the vagus nerve reduced peritoneal IL-6 and intestinal inflammation

Page 88: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

4Va

gal A

nti-i

nflam

mat

ory

Path

way

Med

iate

d th

roug

h N

icot

inic

Jak

-2/S

tat-3

Act

ivat

ion

87

in Stat3fl/+ control mice but failed to do so in LysM-Stat3fl/- mice. These data support the

critical function of STAT3 activation in the cholinergic anti-inflammatory pathway in vivo.

Figure 8. Stimulation of the vagus nerve activates STAT3 in intestinal macrophages in muscularis tissue. (see fullcolor chapter 11)Cholinergic nerve fibers are in close anatomical apposition to macrophages in small intestine. (a) Confocal microscopy of macrophages (ED2; red) and cholinergic nerve fibers (vesicular acetylcho-line transporter; green) around the myenteric plexus of rat ileum. Arrows indicate close anatomi-cal appositions of varicose cholinergic nerve fibers and macrophages at the perimeter of myenteric ganglia and the tertiary plexus outside the ganglia (arrowheads). Scale bar, 10 μm. (b) Mouse ileum sections stained for phosphorylated STAT3 1 h after control laparotomy surgery (L sham), intestinal manipulation (IM sham) or intestinal manipulation combined with stimulation of the vagus nerve (IM

VNS). Transverse section of a complete ileal villus of a control mouse (control laparotomy). SM, submucosa; CM, circular muscle layer; LM, longitudinal muscle layer; MP, myenteric plexus. Arrowheads indicate phosphorylated STAT3−positive nuclei. Scale bar, 20 μm (40 μm for left image). (c) Phos-phorylated STAT3−positive nuclei (green) in mouse ileum 1 h after intestinal manipulation plus stimulation of the vagus nerve, visualized by confocal microscopy. Arrowheads indi-cate colocalization of phosphorylated STAT3 nuclei (PYS-TAT3; green) with phagocytes prelabeled by prior injection of Alexa 546−labeled dextran particles (red). Nuclear coun-terstain is 4,6-diamidino-2-phenylindole (DaPi; blue). Inset, enlarged macrophage showing dextran particles and STAT3 immunoreactivity. Scale bar, 20 μm (10 μm for boxed area). Experiments are representative of three independent incuba-tions in three mice per group.

Page 89: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

88

IL-6

(pg/

ml)

LysM-STAT3fl/–

0

300

600*

LysM-STAT3fl/–

0

500

1,000 *

*

Cel

l den

sity

(cel

ls/m

m2 )*

*

Lsham

IMsham

IMVNS

*

Lsham

IMsham

IMVNS

A B

Figure 9. Stimulation of the vagus nerve fails to reduce inflammation in LysM-Stat3fl/- mice.IL-6 was measured in peritoneal lavage fluid (a) and myeloperoxidase-positive inflammatory infil-trates were quantified in muscularis tissues (b) of Stat3fl/+ control mice (filled bars) or LysM-Stat3fl/- mice (open bars) treated with control laparotomy surgery (L sham), intestinal manipulation (IM sham) or intestinal manipulation plus stimulation of the vagus nerve (IM VNS). (a) Peritoneal lavage fluid was collected 3 h after the procedures. (b) Infiltrates were quantified in whole-mount preparations 24 h after the surgical procedures. Data are mean ± s.e.m.; n = 3−4. *, P < 0.05, compared with the respective control laparotomy surgery group (Mann-Whitney U test).

Discussion The cholinergic anti-inflammatory pathway represents a physiological system for controlling

macrophage activation and inflammation in sepsis models1. Its working mechanism

ultimately involves the prevention of NF-κB p65 activity3 after α7 nAChR activation4, but

the exact cellular mechanism has remained unclear. Here we have demonstrated that

nicotine acts on macrophages via the recruitment of Jak2 to the α7 nAChR and activation

of Jak2, thereby initiating the anti-inflammatory STAT3 and SOCS3 signaling cascade.

Notably, recruitment of Jak2 to the α7 nAChR subunit has also been described in neuronal

PC12 cells exposed to nicotine, as part of a neuroprotective mechanism against β-amyloid-

induced apoptosis20. Our results in resident peritoneal macrophages were consistent with

our in vivo data, as we found activation of STAT3 in intestinal macrophages in response

to stimulation of the vagus nerve in mice, which indicates activation of STAT3 induced by

acetylcholine derived from vagal efferents.

Page 90: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

4Va

gal A

nti-i

nflam

mat

ory

Path

way

Med

iate

d th

roug

h N

icot

inic

Jak

-2/S

tat-3

Act

ivat

ion

89

Activation of the STAT3 cascade after nAChR ligation is fully consistent with the observed

inhibition of proinflammatory cytokine release by macrophages, because STAT3 is a

negative regulator of the inflammatory response6, 27. In our studies, the anti-inflammatory

effect of nicotine on macrophages required DNA binding and transactivation of STAT3, as

nicotine failed to inhibit TNF production in macrophages overexpressing STAT3 altered

in its in DNA-binding capacity17. Likewise, activation of STAT3 is required for the anti-

inflammatory properties of IL-108,28 and the IL-10-induced attenuation of cytokine production

and proliferation28. In addition, STAT3 phosphorylation is required for IL-6-induced growth

arrest and differentiation29.

SOCS3 specifically disables STAT3 phosphorylation via IL-6R but does not interfere with

IL-10R signaling9, 10, 30. Conditional knockout mice specifically lacking SOCS3 in their

macrophages (LysM-Socs3fl/-) show resistance to endotoxemia, explained by the anti-

inflammatory effect of sustained STAT3 activation through IL-6R ligands10. Regardless

of that finding, our results have indicated that the enhanced expression of SOCS3 did

not contribute to the anti-inflammatory effect of nAChR activation, as blockade of SOCS3

expression did not prevent the anti-inflammatory action of nicotine. Hence, the anti-

inflammatory effect of cholinergic activation in macrophages rests mainly on enhanced

STAT3 rather than SOCS3 activation.

We have shown that STAT3 was activated by nicotine directly and that involvement of

enhanced signaling via IL-10R here was unlikely, as we found the macrophage deactivation

induced by stimulation of the vagus nerve to be similarly effective in IL-10-deficient mice.

Moreover, nicotine-induced STAT3 activation could be prevented by nAChR blockers.

Our observations suggest that the molecular route exerting the anti-inflammatory effect

of nAChR activation mimics the signaling pathway of IL-10R without the requirement of

IL-10 itself. That hypothesis is supported by our finding and those of another study2 that,

consistent with the action of IL-1031, nicotine does not alter TNF mRNA expression but

decreases the release of TNF protein. Furthermore, LysM-Stat3fl/- mice have a phenotype

resembling that of IL-10-deficient mice6. Nicotine-induced inhibition of the release of high-

mobility group box 1 in mouse RAW264.7 macrophages is associated with inhibition of

NF-κB p65 transcriptional activity3. Our finding that nicotine repressed macrophage activity

Page 91: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

90

via STAT3 may very well explain that observation, as IL-10−STAT328 signaling blocks NF-

κB DNA-binding32, 33, possibly through direct interaction of dimerized STAT3 with the p65

subunit34.

We have shown here that recruitment of inflammatory infiltrates induced by bowel

manipulation and the resulting symptoms of postoperative ileus were reduced substantially

by stimulation of the vagus nerve. Our results have shown strict cholinergic control of

macrophage activation in vivo, which may be substantiated by the observation that

cholinergic (vesicular acetylcholine transporter−positive) nerve fibers are situated in close

proximity to resident macrophages in intestinal myenteric plexus. At first glance, our data

may seem contradictory to the outcome of earlier attempts to treat postoperative ileus

using cholinergic agents such as neostigmine, which had only limited success35. That lack

of efficacy could be explained by the fact that the inflammatory process had already been

fully accomplished by the time these agents were administered, leaving the activation of

inhibitory neural pathways11 unaffected. Our results indicate that nicotinic receptor activation

before or during surgery prevents postoperative intestinal inflammation and will certainly be

a promising strategy for treating postoperative ileus. Notably, vagus nerve stimulators are

clinically approved devices for the treatment of epilepsy and depression36. In conclusion,

we have shown here that inhibition of macrophage activation via the cholinergic anti-

inflammatory pathway is brought about via Jak2-STAT3 signaling. Our data may aid in

further development of therapeutic strategies for modifying the cholinergic anti-inflammatory

pathway to treat various inflammatory conditions.

Page 92: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

4Va

gal A

nti-i

nflam

mat

ory

Path

way

Med

iate

d th

roug

h N

icot

inic

Jak

-2/S

tat-3

Act

ivat

ion

91

Reference ListTracey, K.J. The inflammatory reflex. Nature 420, 853–859 (2002). 1. Borovikova, L.V. et al. Vagus nerve stimulation attenuates the systemic inflammatory response 2. to endotoxin. Nature 405, 458–462 (2000). Wang, H. et al. Cholinergic agonists inhibit HMGB1 release and improve survival in experimen-3. tal sepsis. Nat. Med. 10, 1216–1221 (2004). Wang, H. et al. Nicotinic acetylcholine receptor α7 subunit is an essential regulator of inflam-4. mation. Nature 421, 384–388 (2003). Matsunaga, K., Klein, T.W., Friedman, H. & Yamamoto, Y. Involvement of nicotinic acetylcholine 5. receptors in suppression of antimicrobial activity and cytokine responses of alveolar mac-rophages to Legionella pneumophila infection by nicotine. J. Immunol. 167, 6518–6524 (2001). Takeda, K. et al. Enhanced Th1 activity and development of chronic enterocolitis in mice devoid 6. of Stat3 in macrophages and neutrophils. Immunity 10, 39–49 (1999). Welte, T. et al. STAT3 deletion during hematopoiesis causes Crohn’s disease-like pathogen-7. esis and lethality: a critical role of STAT3 in innate immunity. Proc. Natl. Acad. Sci. USA 100, 1879–1884 (2003). Levy, D.E. & Lee, C.K. What does Stat3 do? J. Clin. Invest. 109, 1143–1148 (2002). 8. Kubo, M., Hanada, T. & Yoshimura, A. Suppressors of cytokine signaling and immunity. Nat. 9. Immunol. 4, 1169–1176 (2003). Yasukawa, H. et al. IL-6 induces an anti-inflammatory response in the absence of SOCS3 in 10. macrophages. Nat. Immunol. 4, 551–556 (2003). de Jonge, W.J. et al. Postoperative ileus is maintained by intestinal immune infiltrates that acti-11. vate inhibitory neural pathways in mice. Gastroenterology 125, 1137–1147 (2003). Kalff, J.C. et al. Intra-abdominal activation of a local inflammatory response within the human 12. muscularis externa during laparotomy. Ann. Surg. 237, 301–315 (2003). Livingston, E.H. & Passaro, E.P., Jr. Postoperative ileus. Dig. Dis. Sci. 35, 121–132 (1990). 13. Bauer, A.J. & Boeckxstaens, G.E. Mechanisms of postoperative ileus. Neurogastroenterol. 14. Motil. 16, 54–60 (2004). Mikkelsen, H.B., Mirsky, R., Jessen, K.R. & Thuneberg, L. Macrophage-like cells in muscularis 15. externa of mouse small intestine: immunohistochemical localization of F4/80, M1/70, and Ia-antigen. Cell Tissue Res. 252, 301–306 (1988). Shimozaki, K., Nakajima, K., Hirano, T. & Nagata, S. Involvement of STAT3 in the granulocyte 16. colony-stimulating factor-induced differentiation of myeloid cells. J. Biol. Chem. 272, 25184–25189 (1997). de Koning, J.P. et al. STAT3-mediated differentiation and survival and of myeloid cells in re-17. sponse to granulocyte colony-stimulating factor: role for the cyclin-dependent kinase inhibitor p27(Kip1). Oncogene 19, 3290–3298 (2000). Lukas, R.J. et al. International Union of Pharmacology. XX. Current status of the nomenclature 18. for nicotinic acetylcholine receptors and their subunits. Pharmacol. Rev. 51, 397–401 (1999). Meydan, N. et al. Inhibition of acute lymphoblastic leukaemia by a Jak-2 inhibitor. Nature 379, 19. 645–648 (1996). Shaw, S., Bencherif, M. & Marrero, M.B. Janus kinase 2, an early target of alpha 7 nicotinic 20. acetylcholine receptor-mediated neuroprotection against A β-(1–42) amyloid. J. Biol. Chem. 277, 44920–44924 (2002). Drisdel, R.C. & Green, W.N. Neuronal α-bungarotoxin receptors are alpha7 subunit homomers. 21. J. Neurosci. 20, 133–139 (2000). Kalff, J.C. et al. Surgically induced leukocytic infiltrates within the rat intestinal muscularis medi-22. ate postoperative ileus. Gastroenterology 117, 378–387 (1999). de Jonge, W.J. et al. Mast cell degranulation during abdominal surgery initiates postoperative 23. ileus in mice. Gastroenterology 127, 535–545 (2004). Takahashi, T. & Owyang, C. Vagal control of nitric oxide and vasoactive intestinal polypeptide 24. release in the regulation of gastric relaxation in rat. J. Physiol. (Lond.) 484, 481–492 (1995).

Page 93: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

92

Ozaki, H. et al. Isolation and characterization of resident macrophages from the smooth muscle 25. layers of murine small intestine. Neurogastroenterol. Motil. 16, 39–51 (2004). Mikkelsen, H.B. Macrophages in the external muscle layers of mammalian intestines. Histol. 26. Histopathol. 10, 719–736 (1995). Wang, T. et al. Regulation of the innate and adaptive immune responses by Stat-3 signaling in 27. tumor cells. Nat. Med. 10, 48–54 (2004). Williams, L.M., Ricchetti, G., Sarma, U., Smallie, T. & Foxwell, B.M. Interleukin-10 suppression 28. of myeloid cell activation - a continuing puzzle. Immunology 113, 281–292 (2004 . Akira, S. et al. Molecular cloning of APRF, a novel IFN-stimulated gene factor 3 p91-related 29. transcription factor involved in the gp130-mediated signaling pathway. Cell 77, 63–71 (1994). Lang, R. et al. SOCS3 regulates the plasticity of gp130 signaling. Nat. Immunol. 4, 546–550 30. (2003). Kontoyiannis, D. et al. Interleukin-10 targets p38 MAPK to modulate ARE-dependent TNF 31. mRNA translation and limit intestinal pathology. EMBO J. 20, 3760–3770 (2001). Wang, P., Wu, P., Siegel, M.I., Egan, R.W. & Billah, M.M. Interleukin (IL)-10 inhibits nuclear fac-32. tor κB (NF κB) activation in human monocytes. IL-10 and IL-4 suppress cytokine synthesis by different mechanisms. J. Biol. Chem. 270, 9558–9563 (1995). Schottelius, A.J., Mayo, M.W., Sartor, R.B. & Baldwin, A.S., Jr. Interleukin-10 signaling blocks 33. inhibitor of κB kinase activity and nuclear factor κB DNA binding. J. Biol. Chem. 274, 31868–31874 (1999). Yu, Z., Zhang, W. & Kone, B.C. Signal transducers and activators of transcription 3 (STAT3) 34. inhibits transcription of the inducible nitric oxide synthase gene by interacting with nuclear fac-tor κB. Biochem. J. 367, 97–105 (2002). Longo, W.E. & Vernava, A.M., III. Prokinetic agents for lower gastrointestinal motility disorders. 35. Dis. Colon Rectum 36, 696–708 (1993). George, M.S. et al. Vagus nerve stimulation. A potential therapy for resistant depression? Psy-36. chiatr. Clin. North Am. 23, 757–783 (2000). Bennink, R.J. et al. Validation of gastric-emptying scintigraphy of solids and liquids in mice us-37. ing dedicated animal pinhole scintigraphy. J. Nucl. Med. 44, 1099–1104 (2003). Ramakers, C., Ruijter, J.M., Deprez, R.H. & Moorman, A.F. Assumption-free analysis of quanti-38. tative real-time polymerase chain reaction (PCR) data. Neurosci. Lett. 339, 62–66 (2003).

Page 94: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

4Va

gal A

nti-i

nflam

mat

ory

Path

way

Med

iate

d th

roug

h N

icot

inic

Jak

-2/S

tat-3

Act

ivat

ion

93

Page 95: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

5

Page 96: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

5Cha

pter

5Activation of the Cholinergic

Anti-Inflammatory Pathway

Ameliorates Postoperative

Ileus in Mice

Frans O. The, Guy E. Boeckxstaens,

Susanne A. Snoek, Jenna L. Cash,

Roelof J. Bennink, Gregory J. Larosa,

René M. van den Wijngaard, David R. Greaves,

Wouter J. de Jonge

Gastroenterolgy 2007; 133: 1219-1228

Page 97: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

96

AbstractBackground & Aims: We previously showed that intestinal inflammation is reduced by

electrical stimulation of the efferent vagus nerve, which prevents postoperative ileus in

mice. We propose that this cholinergic anti-inflammatory pathway is mediated via alpha7

nicotinic acetylcholine receptors expressed on macrophages. The aim of this study was to

evaluate pharmacologic activation of the cholinergic anti-inflammatory pathway in a mouse

model for postoperative ileus using the alpha7 nicotinic acetylcholine receptor-agonist AR-

R17779. Methods: Mice were pretreated with vehicle, nicotine, or AR-R17779 20 minutes

before a laparotomy (L) or intestinal manipulation (IM). Twenty-four hours thereafter gastric

emptying was determined using scintigraphy and intestinal muscle inflammation was

quantified. Nuclear factor-κB transcriptional activity and cytokine production was assayed

in peritoneal macrophages. Results: Twenty-four hours after surgery IM led to a delayed

gastric emptying compared with L (gastric retention: L + saline 14% ± 4% vs IM + saline

38% ± 10%, P = 0.04). Pretreatment with AR-R17779 prevented delayed gastric emptying

(IM + AR-R17779 15% ± 4%, P = 0.03). IM elicited inflammatory cell recruitment (L + saline

50 ± 8 vs IM + saline 434 ± 71 cells/mm2, P = 0.001) which was reduced by AR-R17779

pretreatment (IM + AR-R17779 231 ± 32 cells/mm2, P = 0.04). An equimolar dose of nicotine

was not tolerated. Subdiaphragmal vagotomy did not affect the anti-inflammatory properties

of AR-R17779. In peritoneal macrophages, both nicotinic agonists reduced nuclear factor

κB transcriptional activity and proinflammatory cytokine production, with nicotine being

more effective than AR-R17779. Conclusions: AR-R17779 treatment potently prevents

postoperative ileus, whereas toxicity limits nicotine administration to ineffective doses.

Our data further imply that nicotinic inhibition of macrophage activation may involve other

receptors in addition to alpha7 nicotinic acetylcholine receptor.

Page 98: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

5Pe

riphe

ral N

icot

inic

Ago

nist

s A

mel

iora

te P

osto

pera

tive

Ileus

97

PBackgroundPostoperative ileus (POI) is characterized by impaired propulsive function of the entire

gastrointestinal tract after abdominal surgery.1 Although normal peristalsis is restored after

3–5 days, POI inflicts patient discomfort (eg, nausea, vomiting, abdominal pain), accounts

for a considerable increase in morbidity, and prolongs hospitalization.2 The additional

annual health care expenses related to POI in the United States are estimated to exceed

1 billion dollars.1,2

Research in rodent models for this pathologic condition has revealed that handling of

the intestine during abdominal surgical procedures initiates a biphasic response. Initially,

spinal and supraspinal inhibitory pathways become activated, enhancing central release

of corticotrophin- releasing factor.3,4 This sympathetic stress response results in the

instantaneous impairment of gastric emptying, lasting up to 3 hours.4 An inflammatory

response of the muscularis propria mediates the delay in gastrointestinal transit observed

up to 24 hours thereafter and represents the prolonged phase in POI.5–7 The importance

of this induced intestinal leukocyte infiltration in the pathogenesis of POI is stressed by the

observation that prevention of this inflammation ameliorates POI.5 Intercellular adhesion

molecule 1 targeting antibodies or antisense oligonucleotides prevent extravasation of

leukocytes to the intestinal muscle layer and normalize gastric emptying, indicating a

reduced POI.6 Activation of macrophages that reside in the intestinal muscle layer have

been implicated to play an important role in the initiation of the manipulation-induced muscle

inflammation.7 Recently, the vagus nerve has been put forward to represent an inhibitory

feedback mechanism that negatively regulates innate immune responses.8,9 Enhanced

efferent vagal nerve output has been shown to reduce inflammatory responses in rodent

models for sepsis, ischemia/ reperfusion, pancreatitis, and POI.10–13 Its anti-inflammatory

potency most likely involves activation of the nicotinic acetylcholine receptors (nAChRs)

on immune cells such as macrophages.10,13,14 The cellular pathways of nicotinic inhibition

of macrophage activation involves the anti-inflammatory Janus kinase 2 (Jak2)/signal

transducer and activator of transcription 3 (STAT3) signaling pathway10 and inhibition of

nuclear factor κB (NF-κB) signaling.15

Page 99: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

98

Previous studies have indicated that nicotine has anti-inflammatory properties. Ghia et al16

recently showed an important role for cholinergic inflammatory control in 2 experimental

colitis models. Chemical as well as surgical blockade of vagal nerve signaling results in a

significant increase of inflammation. Conversely, nicotine treatment resulted in reduction

of the inflammatory response, independent of vagal nerve activity. However, even though

some clinical studies evaluating the role of nicotine in inflammatory bowel disease show

improvement compared with placebo, results generally are disappointing and administration

provokes significant toxic adverse events.17 Given the purported role of alpha7 nAChRs in

mediating the cholinergic anti-inflammatory pathway, 8,10,14 specific alpha7 nAChR agonists

may have higher therapeutic potential than general nicotinic agonists. Because of the

growing interest in manipulation of central nAChRs to treat neuropsychologic disorders

such as Alzheimer’s disease, attention deficit hyperactivity disorder, and schizophrenia,

several of such agonists have been developed in the past decade.18 The nAChR agonist

AR-R17779, a spirooxazolidinone, has a high affinity for the alpha7 receptor subtype19 and

potently activates peripheral as well as central alpha7 nAChRs.19–22

In the present study, we show that the anti-inflammatory efficacy attained with electrical

vagal nerve stimulation can be mimicked by AR-R17779. Pretreatment with AR-R17779

ameliorates POI and reduces the manipulation-induced inflammatory response, although

a similar treatment with nicotine is ineffective. On the other hand, although both nicotinic

agonists reduced activation of peritoneal macrophages, nicotine was more potent in

reducing cytokine release and NF-κB activation as compared with AR-R17779.

Page 100: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

5Pe

riphe

ral N

icot

inic

Ago

nist

s A

mel

iora

te P

osto

pera

tive

Ileus

99

Materials and MethodsReagents and AntibodiesNicotine used ([-]-nicotine) and Zymosan A from S. cerevisiae, was from Sigma-Aldrich

(Zwijndrecht, the Netherlands). Antibodies against nAChR alpha7 were obtained from

Abcam (Cambridge, UK), anti–STAT-3 (PY705) from Cell Signaling Technology (Beverly,

MD), and goat polyclonal anti–β-actin, rabbit polyclonal anti–STAT-3 were from Santa Cruz

Biotechnology, Inc (Santa Cruz, CA). Rat monoclonal anti-F4/80 was obtained from Serotec

(Oxford, UK). The enzyme-linked immunosorbent assays for interleukin-6, Keratinocyl-

derived chemokins (KC), tumor necrosis factor (TNF), and RANTES were purchased from

R&D Systems (Minneapolis, MN).

AnimalsFemale Balb/C mice (Harlan Nederland, Horst, The Netherlands), 12–15 weeks, were kept

under environmentally controlled conditions (light on from 8:00 AM until 8:00 PM; water

and rodent nonpurified diet ad libitum; temperature, 20°C–22°C; humidity, 55%). LysMCre

and STAT-3flox/flox mice23 were kindly made available by Dr S. Uematsu and Professor S.

Akira (Osaka University, Osaka, Japan). All experiments were performed according to the

guidelines of the Ethical Animal Research Committee of the University of Amsterdam.

Study ProtocolMice were assigned randomly to 1 of 7 treatment groups (ie, sham [stimulation], 5 V electrical

vagus nerve stimulation, vehicle [saline], nicotine at 0.9 or 23.0 µmol/kg or AR-R17779 at

0.09, 0.9, or 23.0 µmol/kg). The assigned therapy was administered via intraperitoneal

injection 20 minutes before the surgical procedure was performed as described in the next

section. A subgroup of animals underwent a subdiaphragmal vagotomy 30 minutes before

treatment with saline or AR-R17779.

Surgical ProceduresMice were anesthetized by intraperitoneal injection of a mixture of Fentanyl Citrate/

Fluanisone (Hypnorm; Janssen, Beerse, Belgium) and Midazolam (Dormicum; Roche,

Mijdrecht, The Netherlands). The surgical procedure was performed under sterile conditions.

Page 101: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

100

Mice underwent a laparotomy (L) or small intestinal manipulation (IM) as described

previously.5 In short, the small intestine was exteriorized carefully and manipulated gently

for 5 minutes using sterile, moist cotton applicators. After repositioning of the intestinal

loops, the abdomen was closed using a 2-layer continuous suture (Syneture Sofsilk 6-0).

Mice recovered from surgery in a temperature-controlled cage at 32°C with free access to

water, but not food. Twenty-four hours after surgery, gastric emptying was measured by a

scintigraphic method23 and mice were killed by cervical dislocation. The small intestine was

removed, flushed in ice-cold phosphate-buffered saline (PBS), and snap-frozen in liquid

nitrogen or fixed in ethanol 100% for 10 minutes and stored in ethanol 70% at 4°C until

further analysis.

Electrical Vagal Nerve StimulationElectrical vagal nerve stimulation (EVNS) was performed as described previously.10 To

minimize cardiovascular responses, the left cervical branch was stimulated, avoiding

sinoatrial-induced bradycardia. Five-volt stimuli with a frequency of 5 Hz, 5 ms10 were

applied for 5 minutes before and 15 minutes after abdominal surgery. For sham stimulation,

a cervical midline incision was made, after which the wound was covered with sterile, moist

gauzes for 20 minutes.

Subdiaphragmal VagotomyA midline incision was made under general anesthesia, after which a retractor was placed.

Under microscopic view, both vagal nerve trunks were cut, distal from the diaphragm but

proximal to the division of the hepatic branch. During this procedure, the intraperitoneal

organs were protected and kept moist using sterile gauzes drenched in 0.9% NaCl. Any

palpation or manipulation of the small intestine was specifically avoided. The abdomen

was closed using a 2-layer continuous suture (Syneture Sofsilk 6-0). Animals were kept in

a temperaturecontrolled cage at 32°C for 30 minutes, after which they entered the study

protocol.

Measurement of Gastric EmptyingGastric emptying was determined as described previously.24 After gavage of a semiliquid,

noncaloric test meal (0.1 mL of 3% methylcellulose solution containing 10 MBq of 99mTc-

Albures), mice were scanned using a gamma camera set at 140 keV.24 The entire abdominal

Page 102: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

5Pe

riphe

ral N

icot

inic

Ago

nist

s A

mel

iora

te P

osto

pera

tive

Ileus

101

region was scanned for 30 seconds, immediately and 80 minutes after gavage.6,24 During

the scanning period mice were conscious and restrained manually. The static images

obtained were analyzed using Hermes computer software (Hermes, Stockholm, Sweden).

Gastric retention was calculated by determining the percentage of activity present in the

gastric region of interest compared with the total abdominal region of interest.6,24

Whole-Mount PreparationAs previously described,5 the mucosa was separated carefully from the muscle layer. Fixed

preparations were rehydrated by incubation in 50% ethanol and PBS, pH 7.4, for 5 minutes.

To visualize myeloperoxidasepositive cells preparations were incubated for 10 minutes with

3-amino-9-ethyl carbazole (Sigma, St. Louis, MO) as a substrate and dissolved in sodium

acetate buffer (pH 5.0) to which 0.01% H2O2 was added.5 To quantify the extent of intestinal

muscle inflammation, the number of myeloperoxidase-positive cells in 3 randomly chosen

1-mm2 fields were counted and expressed as the number of myeloperoxidase-positive cells

per mm2.

Cell Culture and ImmunohistochemistryResident peritoneal macrophages were harvested by flushing the peritoneal cavity with

5 mL of Hank’s balanced salt solution containing 10 U/mL heparin. Peritoneal cells were

plated in Opti-Mem I medium (Gibco, Carlsbad, CA), supplemented with 10 mmol/L

L-glutamine, 100 U/mL penicillin, and 100 µg/mL gentamycingentamycin. Macrophages

were left to adhere for 2 hours in a humidified atmosphere at 37°C with 5% CO2. Cells were

washed and adhering cells were left for 16–20 hours. Subsequently, cells were stimulated

with lipopolysaccharide (LPS) (Escherichia coli 100 ng/mL; Sigma-Aldrich) and interferon-γ

(10 ng/mL) in the presence of indicated concentrations of nicotinic agonist for 3 hours, or

lysed 30 minutes after nicotinic agonist/LPS/interferon-γ exposure for immunoblotting, as

described.10 For confocal microscopy, macrophages were left to adhere for 16–20 hours

on glass slides (Nunc, Rochester, NY) in RPMI medium supplemented with 10% fetal calf

serum. Cells were washed 5 times with ice-cold Hank’s balanced salt solution containing 1

mmol/L Na3VO4, and fixed in ice-cold 4% phosphate-buffered (pH 7.4) paraformaldehyde

for 1 hour. After washing with ice-cold PBS pH 7.4, cells were stained with appropriate

antibodies at 4°C for 16–20 hours. Antibodies were visualized using anti-rat Alexa546-

labeled secondary antibodies and biotinlabeled anti-rabbit antibodies, followed by Alexa

Page 103: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

102

488-streptavidin (Molecular Probes). Sections were mounted in glycerol mounting medium

to which DAPI (10 µg/mL; Molecular Probes) nuclear counter stain was added.

ImmunoblottingAs described,10 cells were scraped in 50 µL of ice-cold lysis buffer containing 150 mmol/L

NaCl, 0.5% Triton X-100, 5 mmol/L ethylenediaminetetraacetic acid, 0.1% sodium dodecyl

sulfate, 0.5% deoxycholate, 10% glycerol, 1 mmol/L Na3VO4, 50 mmol/L NaF, 1 µg/mL

aprotinin, 1 µg/mL leupeptin, and 1 mmol/L phenylmethylsulfonyl fluoride. Samples were

suspended in 50µL sample buffer (125 mmol/L Tris-HCl, pH 6.8, 2% sodium dodecyl

sulfate, 10% β-mercaptoethanol, 10% glycerol, and 0.5 mg/mL bromophenol blue),

loaded onto sodium dodecyl sulfate–polyacrylamide gel electrophoresis gels, and blotted

onto polyvinylidene difluoride membranes (Millipore). Membranes were blocked in Tris-

buffered saline/0.1% Tween-20 containing 5% nonfat dry milk and incubated overnight with

appropriate antibodies in Tris-buffered saline/0.1% Tween-20/1% bovine serum albumin.

Horseradish-peroxidase–conjugated secondary antibodies were visualized using Lumilite

plus (Boehringer-Mannheim, Germany).

Reverse-Transcription Polymerase Chain ReactionTotal RNA from tissue was isolated using Trizol (Invitrogen, Carlsbad, CA), treated with DNase,

and reverse transcribed. The resulting complementary DNA (0.5ng) was subjected to Light

Cycler polymerase chain reaction (CYBR Green Fast start polymerase; Roche, Mannheim,

Germany) for 40 cycles. Primers used were TNFα forward 5’ GACAAGGCTGCCCCGACTA

3’; reverse 5’AGGAGGTTGACTTTCT CCTGGTATG 3’, and HPRT forward

5’GACCGGTCCCGTCATGC 3’; reverse 5’ TCATA-ACCTGGTTCATCATCGC 3’; RANTES

forward 5’ GACACCACTCCCTGCTGCT 3’, reverse 5’ GAAATACTCCTTGACGTGGGCA

3’.

NF-κB Activity AssayImmortalized splenic macrophages Mf4/425 (a kind gift from Professor M. P. Peppelenbosch,

University of Groningen, Groningen, the Netherlands) were co-transfected with NF-

κB luciferase and cytomegalovirus renilla luciferase reporter constructs (Clontech,

MountainView, CA) using Jet PEI (PolyTransfection), according to the manufacturer’s

instructions. Briefly, 0.5 µg per 106 cells of constructs NF-κB–luc and 5 ng cytomegalovirus

Page 104: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

5Pe

riphe

ral N

icot

inic

Ago

nist

s A

mel

iora

te P

osto

pera

tive

Ileus

103

Renilla Luciferase was suspended in 75 µL of 150 mmol/L sterile NaCl solution. Also,

1.6 µL of Jet PEI solutions was suspended in 75 µL of 150 mmol/L sterile NaCl solution.

The Jet PEI/NaCl solution then was added to the DNA/NaCl solution and incubated at

room temperature for 30 minutes and 150 µL of the DNA/Jet PEI was added to the cells.

The transfection was allowed to proceed for 16 hours, and the medium was refreshed.

Twenty-four hours after transfection, cells were pretreated with nicotinic agonists at the

concentration indicated for 1 hour, and subsequently stimulated with zymosan (5 particles

per cell) for 6 hours. After treatment, the medium was removed; the cells were washed

3 times with ice-cold PBS, the cells were lysed with Passive Lysis Buffer supplied in the

Dual Luciferase Reporter Assay Kit (Promega), and the lysate was assayed for luciferase

activity according to the manufacturer’s instructions.

StatisticsStatistical analysis was performed with the use of SPSS 12.02 software for Windows

(SPSS, Inc, Chicago, IL). Data were analyzed using the nonparametric Mann–Whitney

U test for independent samples. The Friedman’s 2-way analysis of variance was used to

explore multiple dependent value assays (ie, the in vitro inflammatory protein response).

If the Friedman’s analysis was significant, individual values compared with the 0-nmol/L

concentration were tested with a Mann–Whitney U test. P values less than 0.05 were

considered statistically significant and results were depicted as mean ± SEM.

Page 105: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

104

ResultsElectrical Vagus Nerve Stimulation and AR-R17779 Prevent POIConsistent with our previous results,5,10 gastric emptying was impaired significantly in mice

subjected to IM when compared with L alone (Figure 1A). First, we confirmed that activation

of the cholinergic anti-inflammatory pathway by electrical stimulation of the vagus nerve

during surgery ameliorates POI. The postoperative delay in gastric emptying resulting from

IM was reduced significantly if the surgical procedure was combined with a 5-V electrical

stimulation of the left cervical vagus nerve for 20 minutes (Figure 1A).

L IMsaline

-

rela

tive

10

20

30

40

50

60

gast

ric c

onte

nt (%

) #

##

A

** **

*

L IMEVNS

L IMsham

surgerytreatmentdose (mg/kg)

L IMnicotine

0.4

L IMAR-R0.2

L IMAR-R5.0

B

100

200

300

400

500

600

MPO

-pos

. ce

lls/m

m2

#

**

*

##

L IMEVNS

L IMsham

surgerytreatmentdose (mg/kg)

L IMnicotine

0.4

L IMAR-R0.2

L IMsaline

-

L IMAR-R5.0

L IMAR-R0.02

L IMAR-R0.02

Page 106: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

5Pe

riphe

ral N

icot

inic

Ago

nist

s A

mel

iora

te P

osto

pera

tive

Ileus

105

Gastric retention measured 24 hours after electrical vagal nerve stimulation in L control

mice indicated that electrical vagal nerve stimulation by itself did not influence postoperative

gastric emptying 24 hours after the procedure (Figure 1A sham vs EVNS).

To explore the potential of pharmacologic nAChR activation of the cholinergic anti-

inflammatory pathway, we next investigated whether administration of nAChR agonist

nicotine or alpha7 nAChR agonist AR-R17779 would ameliorate POI. Administration of AR-

R17779 in a dose of 0.2 or 5 mg/kg restored gastric emptying to the levels seen with L alone

(Figure 1A). In contrast, nicotine at a dose equimolar to 0.2 mg/kg AR-R17779 (0.4 mg/kg)

failed to improve gastric emptying (Figure 1A), although nicotine administration at this dose

range has been reported to already cause significant behavioral changes in rats.20 Mice

treated with higher doses of nicotine (up to a dose equimolar to 5 mg/kg AR-R17779; 10.6

mg/kg nicotine) clearly developed behavioral agitation within seconds after administration,

illustrating marked neurotoxicity, in agreement with other studies.26 Therefore, no further

experiments were conducted using nicotine at doses higher than 0.4 mg/kg.

L IM

-

ei 30

t

** **

L I IMurgery

dose ( g/ g)

L M

0.4

L M

2

L M

5

C #*

**

100

200

300

400

500

600

MPO

-pos

. ce

lls/m

m2

L IMVGX

saline

L IMVGX

AR-R 5.0

L IMshamsaline

surgerysham/VGXtreatment

#

B

1

300

0

600

MO

o

cs

#

*

L IM

0 2

Figure 1. POI is ameliorated after treatment with alpha7-selective agonists AR-R17779. (A) Gastric retention 80 minutes after gavage of a semiliquid test meal in mice that had undergone the indicated treatment and L or IM 24 hours previously. (B) Quantitative analysis of IM-induced inflammatory cell recruitment 24 hours after indicated treatment and surgery. (C) Quantitative analysis of manipulation-induced inflammatory cell recruitment 24 hours after indicated treatment and surgery in VGX or sham-vagotomized animals. Data shown are mean values ± SEM of 6–8 mice. L, ; IM, #P < 0.05 vs sham laparotomy. ##vs sham IM. *P < 0.05 vs saline laparotomy. **P < 0.05 vs saline IM.

Page 107: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

106

Manipulation-Induced Inflammation Is Decreased by Electrical Vagal Nerve Stimulation and Pretreatment With AR-R17779We established previously that delayed gastric emptying results from an intestinal muscle

inflammation inflicted by the surgical bowel handling.5 Because electrical vagal nerve

stimulation and AR-R17779 also acts on neuronal receptors we investigated whether

the improved gastric emptying results from inhibition of the inflammatory response in

manipulated muscle tissue. IM initiated a marked recruitment of inflammatory cells to the

muscle layer of the handled small-bowel segment (Figure 1B). However, if IM surgery was

combined with electrical vagal nerve stimulation, the number of inflammatory cells recruited

to the muscle layer was reduced significantly (Figure 1B).

Next, we investigated whether preoperative treatment with nicotinic agonists would attain

a similar anti-inflammatory response. Figure 1B shows that IM results in a significant influx

of leukocytes into the small intestine 24 hours after surgery, whereas pretreatment with

AR-R17779 (5 mg/kg) significantly reduced the number of inflammatory cells infiltrating

the intestinal muscle segment in response to IM. Pretreatment with nicotine 0.4 mg/kg

20

40

60

80

100

0 1 10 1000

Nicotinic agonist (nM)

TNF

% d

ecre

ase

****

**

**

20

40

60

80

100

IL6

% d

ecre

ase

* ** **

0 1 10 1000

Nicotinic agonist (nM)

20

40

60

80

100

KC %

dec

reas

e

0 1 10 1000

Nicotinic agonist (nM)

** ** **

* *

A

AR-R17779

Nicotine

Page 108: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

5Pe

riphe

ral N

icot

inic

Ago

nist

s A

mel

iora

te P

osto

pera

tive

Ileus

107

or its equimolar dose of AR-R17779 (0.2 mg/kg) failed to reduce the number of recruited

inflammatory cells significantly (Figure 1B).

AR-R17779 Pretreatment Reduces Intestinal Muscle Inflammation Independent of Vagal Nerve SignalingOne potential mechanism for the observed anti-inflammatory effects of AR-R17779 could

be activation of central nAChRs and subsequently increased vagal efferent activity. To

investigate whether the effect achieved with AR-R17779 depends on enhanced vagal nerve

signaling, we tested whether AR-R17779 5 mg/kg was effective in mice that had undergone

a subdiaphragmal bilateral vagotomy (VGX) before treatment and IM or L. VGX in itself did

not elicit an intestinal muscle inflammation (L sham vs L VGX, Figure 1C). Importantly, the

potency of AR-R17779 to reduce myeloperoxidase-positive infiltrate recruitment was not

affected by VGX (Figure 1C), indicating that the anti-inflammatory effect of AR-R17779 is

independent of vagal activity.

0 1 10 1000

icotinic agon

%

ec

*

TNF

20

40

60

80

100

*

RANTES

20

40

60

80

100

0 0Vehicle LPS LPS

NicotineLPS

NicotineMLA

ec * *

0 1 1 1000

00

C

dre

e

0 100

**

Vehicle LPS LPSNicotine

LPSNicotine

MLA

Nor

mal

ised

exp

ress

ion

(%)

Nor

mal

ised

exp

ress

ion

(%)

B

R R 79

ico i

Figure 2. Nicotinic agonists reduce cytokine and chemokine production in macrophages. (A) TNF, interleukin-6, or KC release from primary peritoneal macrophages stimulated with LPS (100 ng/mL) in the presence of nicotine ( ) or AR-R17779 ( ) at the indicated concentration. Data shown are mean percentages compared with vehicle/endotoxin treatment baseline concentration ± SEM of 5–7 mice measured in duplicate. *P < 0.05 vs 0 nmol/L nicotine or AR-R17779 baseline concentration.**P < 0.01 vs 0 nmol/L nicotine or AR-R17779 baseline concentration. (B) Nicotine suppresses the up-regulation of inflammatory mediator transcripts by activated macrophages via alpha7 nAChR. Peritoneal macrophages were pretreated with vehicle (media), nicotine (80 nmol/L), or nicotine (80 nmol/L) + methyllycaconitine (MLA; 5mmol/L) for 1 hour and then stimulated with LPS (100 ng/mL) for 15 hours. Cytokine transcript expression was normalized to hypoxanthine phosphoribosyltrans-ferase messenger RNA. Data shown are mean ± SEM from 4 independent experiments using cells from different donors. Asterisks indicate significant differences (P < 0.05) relative to LPS treated samples.

***

*

F NTES

0 1t i (

1 0i i t ( )

** ** *

)

R R 7 7

N co ine

20

80

I

60

80

100

KC

dcr

ese

0 1 10 100

Nicotinic agonist (nM)

* *

*

AR-R1 779

i

Page 109: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

108

Macrophage Activation Is Modulated by Nicotine and AR-R17779We hypothesized that AR-R17779 would exert its anti-inflammatory effect via activation

of peripheral nAChRs on macrophages because nicotinic agonists have been shown

previously to dose-dependently inhibit release of proinflammatory cytokines and chemokines

by macrophages stimulated with endotoxin.10,14 To assess the potency of AR-R17779 to

reduce inflammatory mediator release in vitro, peritoneal macrophages were stimulated

with LPS and interferon-γ in the presence of nicotine or AR-R17779 in a 0–1000 nmol/L

concentration range. As shown in Figure 2A, nicotine as well as AR-R17779 reduced TNF

and KC production in LPS activated macrophages, whereas interleukin-6 (and RANTES,

not shown) was reduced significantly only by nicotine. In conjunction, nicotine inhibited

transcription of TNF and RANTES, an effect that was blocked by a selective alpha7 nAChR

antagonist methyllycaconitine (MLA) (Figure 2B).

Nicotinic agonists have been shown previously to reduce pro-inflammatory cytokine

production via inhibition of NF-κB activation.8 The modest effect of AR-R17779 on pro-

inflammatory mediator production prompted us to explore the potency of AR-R17779 to

reduce NF-κB transcriptional activity. To this end, we investigated the effect of nicotine and

AR-R17779 on NF-κB activation induced by zymosan particles in a reporter assay using

the immortalized splenic macrophage cell line Mf4/4,25 transiently transfected with a κB

responsive element linked to luciferase gene. As shown in Figure 3A, NF-κB transcriptional

activity was induced by zymosan particles. When cells were pretreated with nicotine or

AR-R17779, NF-κB transcriptional activity was reduced. Notably, however, although

nicotine reduced activity to background levels, AR-R17779 failed to reduce NF-κB activity

completely, even at concentrations as high as 10 µmol/L.

We previously reported that nicotinic stimulation of nAChRs on peritoneal macrophages

leads to activation of the Jak2/STAT3 pathway, diminishing its pro-inflammatory cytokine

release.10 We next investigated whether AR-R17779 activated similar pathways in

peritoneal macrophages. Both AR-R17779 and nicotine induced the phosphorylation of

STAT3 in peritoneal macrophages (Figure 4). Immunoblot analysis of cell lysates from

peritoneal macrophages confirmed this observation, showing that nicotine and AR-R17779

led to a dose-dependent increase in STAT3 phosphorylation, although AR-R17779 was less

effective compared with nicotine (Figure 4A). The results of the immunoblot were confirmed

Page 110: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

5Pe

riphe

ral N

icot

inic

Ago

nist

s A

mel

iora

te P

osto

pera

tive

Ileus

109

by immunofluorescent staining of peritoneal macrophages with a phospho-STAT3–specific

antibody (Figure 4B). We earlier reported that in our model for POI, the anti-inflammatory

effect of vagal nerve activation depends on STAT3 activation. We subsequently investigated

whether the anti-inflammatory properties observed with AR-R17779 functionally depend on

STAT3 expression. To this end, we harvested peritoneal macrophages from LysM-Cre/

Stat3flox/- conditional knock-out mice that specifically lack STAT3 in their macrophages and

neutrophils.23 AR-R17779 elicited a dose-dependent reduction in TNF, interleukin-6, and

KC release in endotoxinstimulated peritoneal macrophages from unaffected Stat3flox/flox

control mice, but failed to reduce the release of these cytokines and chemokines in Stat3-

deficient peritoneal macrophages (Figure 5).

0,0

0,2

0,4

0,6

NF-

B ac

tivity

(R

LU lu

cife

rase

/reni

lla)

vehicle Nicotine0.1 μM

Nicotine1.0 μM

AR-R177791.0 μM

AR-R1777910 μM

0,6

0,8

*

**

Figure 3. Nicotinic agonists reduce NF-kB transcriptional activity in macrophages. Macrophages (im-mortalized splenocytes Mf4/4) transiently transfected with a κB firefly luciferase reporter were treated with nicotine or AR-R17779 at the indicated concentrations, and then stimulated with medium ( ) or zymosan (5 particles per cell), respectively; ( ). Cells were co-transfected with a cytomegalovirus–renilla luciferase construct to normalize for transfection efficiency. Shown are normalized means ± SEM of 3–4 independent experiments in duplicate. Asterisks indicate significant differences (P < .05) of LPS vs vehicle.

Page 111: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

110

Figure 4. (see fullcolor chapter 11) Nicotinic agonists induce STAT3 activation in peritoneal mac-rophages. (A) Immunoblots showing a dose-dependent increase of phosphorylated STAT3 in perito-neal macrophages treated with nicotine (0–100 nmol/L) or AR-R17779 (0–1000 nmol/L). Blots shown are representative of 3 independent experiments. (B) Confocal images of peritoneal macrophages attached to glass slides and stimulated with LPS (10 ng/mL) with the addition of either vehicle: AR-R17779 (100 nmol/L), or nicotine (100 nmol/L). Treatment of cells with AR-R17779 (middle) or nicotine (lower) enhances nuclear staining of phosphorylated STAT3 in F4/80 (red)-positive macrophages.

Page 112: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

5Pe

riphe

ral N

icot

inic

Ago

nist

s A

mel

iora

te P

osto

pera

tive

Ileus

111

C

B

A

0 10 100 10000

25

50

75

100 LysMCre/STAT3flox/STAT3flox/flox

AR-R17779 (nM)

TNF-

alph

a%

dec

reas

e

0 10 100 10000

25

50

75

100 LysMCre/STAT3flox/STAT3flox/flox

AR-R17779 (nM)

IL-6

% d

ecre

ase

0 10 100 10000

25

50

75

100 LysMCre/STAT3flox/STAT3flox/flox

AR-R17779 (nM)

KC%

dec

reas

e

*

**

**

**

**

**

Figure 5. (A) TNF, (B) interleukin-6, and (C) KC release by peritoneal macrophages harvested from STAT3flox/flox( ) controls or LysMCre/STAT3flox/flox( ) mice and stimulated with LPS (100 ng/mL) in the presence of nicotine or AR-R17779 (0–1000 nmol/L). Data shown are mean percentages com-pared with vehicle/LPS treatment baseline concentration ± SEM of 6 assays. *P < 0.05 vs 0 nmol/L AR-R17779 baseline concentration. **P < 0.01 vs 0 nmol/L AR-R17779 baseline concentration.

Page 113: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

112

DiscussionThe cholinergic anti-inflammatory pathway is a now well-established mechanism to control macrophage

activation.8,9 Electrical vagal nerve stimulation has been shown to dampen inflammatory responses

via enhanced efferent vagal output.10,13 However, pharmacologic activation of this pathway might be

a more feasible therapeutic strategy to treat a wide range of inflammatory disorders. Here, we have

shown that presurgical systemic administration of the alpha7 nAChR agonist AR-R17779 is effective

in ameliorating POI through the reduction of manipulation-induced inflammation.

The intestinal muscle inflammation resulting from peri-operative bowel handling now is accepted

widely to play an important role in the pathogenesis of prolonged POI.5,7 This self-limiting disturbance

of normal gastrointestinal propulsion inflicts considerable patient discomfort, morbidity, and is a major

cause of prolonged hospitalization.1 Resident intestinal macrophages located between the circular

and longitudinal muscle layer, in close contact with myenteric cholinergic nerve fibers,10 have been

shown to play an important role in the initiation of the manipulation-induced inflammatory response.7

Inhibitory strategies specifically targeting this macrophage population may have potential in the

treatment of POI.

Electrical stimulation of the left cervical vagus nerve reduces manipulation-induced small intestinal

inflammation and prevents the development of POI, consistent with our previous results.10 The

attenuation of macrophage activation achieved by electrical vagal nerve stimulation is mediated by

alpha7 nAChR–dependent STAT3 signaling in intestinal macrophages.10 Therefore, we hypothesized

that pharmacologic interaction with the alpha7 nAChR may embody a noninvasive and attractive

alternative to electrical vagal nerve stimulation. Our results show that systemic single-dose

administration of nicotine in a tolerable dose (although known to already provoke significant toxic

effects20,26) fails to reduce inflammation or improve postoperative gastric emptying. Increasing the

dose revealed striking adverse events (eg, clonic seizure), excluding further experimentation. This

observation is in line with previous nicotine toxicity studies performed in mice26 and the numerous

side effects observed in clinical studies conducted with nicotine in inflammatory bowel disease.27,28 In

contrast, the alpha7-selective agonist AR-R17779 was well tolerated and was effective in reducing

the manipulation-induced inflammation and normalized the gastric emptying rate. The reason why

nicotine fails to reduce inflammation or POI most likely rests on the 5-fold lower affinity and 35,000-

Page 114: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

5Pe

riphe

ral N

icot

inic

Ago

nist

s A

mel

iora

te P

osto

pera

tive

Ileus

113

fold lower selectivity for the alpha7 nAChR of this compound compared with AR-R17779.19 The

absence of an inflammation-dampening response after nicotine treatment in vivo also results, in part,

from the dosing frequency. Ghia et al16 only observed an anti-inflammatory effect after 5 consecutive

days of nicotine administration in drinking water in experimental colitis. Furthermore, their data also

suggested a nicotinic anti-inflammatory effect that was independent of vagal nerve integrity in line

with our current results with AR-R17779.

AR-R17779 does not pass the blood-brain barrier easily, as shown by previous pharmacokinetic

studies. At 30 minutes after intravenous administration of 30 mg/kg AR-R17779 (a dose 6 times

higher than the dose used in the current study) only 7 µmol/L was present in the brain, although the

median effective concentration value for alpha7 nAChR activation is 27 µmol/L.20 Concurrently, VGX

in our present study did not affect the anti-inflammatory potency of AR-R17779. Although AR-R17779

also activates central nAChRs, these data indirectly suggest a peripheral site of action. However,

cerebroventricular organs lack a proper blood-brain barrier and thereby could represent a gateway

to the central nervous system for substances such as AR-R17779. Therefore, a centrally mediated

mechanism, for instance triggering the HPA-axis, cannot be ruled out completely at this time. Microglia,

the central nervous system macrophage, also express alpha7 nAChRs29 and might represent an

alternative central target for AR-R17779. Indeed, stimulation of these receptors results in a reduction

in their LPS-induced TNF release and subsequent neuroinflammation.29 Unexpectedly, VGX did not

lead to a significantly higher inflammatory reaction compared with nonvagotomized animals. Cervical

vagotomy enhances the inflammatory response in models for endotoxemia, pancreatitis, and acute

local inflammation,11,13,30 which has led to the speculation that the cholinergic anti-inflammatory

pathway represents a regulatory mechanism.8,9 On the other hand, Bernik et al31 did not observe a

worsened inflammation after vagotomy in septic shock, nor did Luyer et al32 in hemorrhagic shock. It

may well be that the vagotomy procedure itself elicits the release of acetylcholine, altering the outcome

within a certain time frame. Similar to this study by Luyer et al,32 the interval between vagotomy and

inflammatory insult in our current study was less than 1 hour (45 and 50 min, respectively). In a

different study from our laboratory, subdiaphragmal vagotomy performed more then 1 hour before

intestinal manipulation did elicit a significant increase of intestinal inflammation (unpublished data).

However, the exact explanation remains to be elucidated.

Further analysis of resident peritoneal macrophages showed that exposure to nicotine or AR-

R17779 activates STAT3 signaling and reduces NF-κB activation. These results are in agreement

Page 115: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

114

with previous findings,10,15 and consistent with a model in which the cholinergic anti-inflammatory

pathway is dependent on Jak2/STAT3 signaling downstream from alpha7 nAChR activation through

AR-R17779.10 Hence, a plausible mechanism for the cellular effect of nicotine would be that proteins

in the STAT3 and NF-κB signaling pathways interact to mount an anti-inflammatory response

(ie, as described for p65/c-rel and phosphorylated STAT3).33 This hypothesis is currently under

investigation.

Despite its ability to reduce manipulation-induced inflammatory responses in vivo, AR-R17779 was

less potent in reducing macrophage NF-κB activation and pro-inflammatory mediator release as

compared with nicotine. This implies that the in vivo effects of AR-R17779 in reducing inflammation

may not rest exclusively on the modulation of macrophage activation. Various other cell types involved

in the innate inflammatory response, such as endothelial cells,34 and dendritic cells (unpublished

observation),35 have been shown to express the alpha7 nAChR and may be targeted by AR-R17779.

This is supported further by a recent report that in a rat model of endotoxemia another alpha7 nAChR

agonist, GTS-21, was found to ameliorate endotoxin-induced immune responses by a mechanism

independent of macrophage TNF and MIP2 release.36 In addition, our data indicate that nicotinic

receptors other than alpha7 nAChR may be involved in the effects of macrophage activation

pathways and function in vitro, such as NF-κB activation and cytokine production, consistent with

findings that macrophages express several subtypes of nAChRs.35 In conclusion, we show here that

a single, preoperative dose of the alpha7 agonist AR-R17779 matches the anti-inflammatory potency

of electrical vagal nerve stimulation. AR-R17779 prevents POI in mice and reduces the manipulation-

induced intestinal muscle inflammation. This alpha7 selective agonist binds to its receptor on

macrophages, activating the cholinergic anti-inflammatory pathway in a vagal efferent–independent

manner. Our data encourage further clinical exploration of alpha7-selective agonists such as AR-

R17779 as putative treatment for POI and various other inflammatory disorders involving the innate

immune system.

Page 116: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

5Pe

riphe

ral N

icot

inic

Ago

nist

s A

mel

iora

te P

osto

pera

tive

Ileus

115

Reference ListPrasad M, Matthews JB. Deflating postoperative ileus. Gastroenterology 1999;117:489ñ492.1. Livingston EH, Passaro EP Jr. Postoperative ileus. Dig Dis Sci 1990;35:121ñ132.2. Luckey A, Wang L, Jamieson PM, et al. Corticotropin-releasing factor receptor 1-deficient mice 3. do not develop postoperative gastric ileus. Gastroenterology 2003;125:654ñ659.Barquist E, Bonaz B, Martinez V, et al. Neuronal pathways involved in abdominal surgery-4. induced gastric ileus in rats. Am J Physiol 1996;270:R888ñR894.de Jonge WJ, van den Wijngaard RM, The FO, et al. Postoperative ileus is maintained by 5. intestinal immune infiltrates that activate inhibitory neural pathways in mice. Gastroenterology 2003;125: 1137ñ1147.The FO, de Jonge WJ, Bennink RJ, et al. The ICAM-1 antisense oligonucleotide ISIS-3082 6. prevents the development of postoperative ileus in mice. Br J Pharmacol 2005;146:252ñ258.Kalff JC, Schraut WH, Simmons RL, et al. Surgical manipulation of the gut elicits an intestinal 7. muscularis inflammatory response resulting in postsurgical ileus. Ann Surg 1998;228:652ñ663.Ulloa L. The vagus nerve and the nicotinic anti-inflammatory pathway. Nat Rev Drug Discov 8. 2005;4:673ñ684.Tracey KJ. The inflammatory reflex. Nature 2002;420:853ñ859. 9. de Jonge WJ, van der Zanden EP, The FO, et al. Stimulation of the vagus nerve attenu-10. ates macrophage activation by activating the Jak2-STAT3 signaling pathway. Nat Immunol 2005;6:844ñ851.van Westerloo DJ, Giebelen IA, Florquin S, et al. The vagus nerve and nicotinic receptors 11. modulate experimental pancreatitis severity in mice. Gastroenterology 2006;130:1822ñ1830.Bernik TR, Friedman SG, Ochani M, et al. Cholinergic anti-inflammatory pathway inhibition of 12. tumor necrosis factor during ischemia reperfusion. J Vasc Surg 2002;36:1231ñ1236.Borovikova LV, Ivanova S, Zhang M, et al. Vagus nerve stimulation attenuates the systemic 13. inflammatory response to endotoxin. Nature 2000;405:458ñ462.Wang H, Yu M, Ochani M, et al. Nicotinic acetylcholine receptor alpha7 subunit is an essential 14. regulator of inflammation. Nature 2003;421:384ñ388.Wang H, Liao H, Ochani M, et al. Cholinergic agonists inhibit HMGB1 release and improve 15. survival in experimental sepsis. Nat Med 2004;10:1216ñ1221.Ghia JE, Blennerhassett P, Kumar-Ondiveeran H, et al. The vagus nerve:a tonic inhibitory 16. influence associated with inflammatory bowel disease in a murine model. Gastroenterology 2006;131:1122ñ1130.Thomas GA, Rhodes J, Mani V, et al. Transdermal nicotine as maintenance therapy for ulcer-17. ative colitis. N Engl J Med 1995;332:988ñ992.Levin ED, Rezvani AH. Development of nicotinic drug therapy for cognitive disorders. Eur J 18. Pharmacol 2000;393:141ñ146.Mullen G, Napier J, Balestra M, et al. (-)-Spiro[1-azabicyclo[2.2.2]octane-3,5í-oxazolidin-2í-19. one], a conformationally restricted analogue of acetylcholine, is a highly selective full agonist at the alpha 7 nicotinic acetylcholine receptor. J Med Chem 2000;43:4045ñ4050.Grottick AJ, Trube G, Corrigall WA, et al. Evidence that nicotinic alpha(7) receptors are not 20. involved in the hyperlocomotor and rewarding effects of nicotine. J Pharmacol Exp Ther 2000;294:1112ñ1119.Papke RL, Porter Papke JK, Rose GM. Activity of alpha7-selective agonists at nicotinic 21. and serotonin 5HT3 receptors expressed in Xenopus oocytes. Bioorg Med Chem Lett 2004;14:1849ñ1853.Van Kampen M, Selbach K, Schneider R, et al. AR-R 17779 improves social recognition in rats 22. by activation of nicotinic alpha7 receptors. Psychopharmacology (Berl) 2004;172:375ñ383.Takeda K, Clausen BE, Kaisho T, et al. Enhanced Th1 activity and development of chronic 23. enterocolitis in mice devoid of Stat3 in macrophages and neutrophils. Immunity 1999;10:39ñ49.

Page 117: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

116

Bennink RJ, de Jonge WJ, Symonds EL, et al. Validation of gastric-emptying scintigraphy of 24. solids and liquids in mice using dedicated animal pinhole scintigraphy. J Nucl Med 2003;44: 1099ñ1104.Desmedt M, Rottiers P, Dooms H, et al. Macrophages induce cellular immunity by activating 25. Th1 cell responses and suppressing Th2 cell responses. J Immunol 1998;160:5300ñ5308.Matta SG, Balfour DJ, Benowitz NL, et al. Guidelines on nicotine dose selection for in vivo 26. research. Psychopharmacology (Berl) 2006.Pullan RD, Rhodes J, Ganesh S, et al. Transdermal nicotine for active ulcerative colitis. N Engl 27. J Med 1994;330:811ñ815.Sandborn WJ. Nicotine therapy for ulcerative colitis: a review of rationale, mechanisms, phar-28. macology, and clinical results. Am J Gastroenterol 1999;94:1161ñ1171.Suzuki T, Hide I, Matsubara A, et al. Microglial alpha7 nicotinic acetylcholine receptors drive a 29. phospholipase C/IP3 pathway and modulate the cell activation toward a neuroprotective role.J Neurosci Res 2006;83:1461ñ1470. Borovikova LV, Ivanova S, Nardi D, et al. Role of vagus nerve signaling in CNI-1493-mediated 30. suppression of acute inflammation. Auton Neurosci 2000;85:141ñ147.Bernik TR, Friedman SG, Ochani M, et al. Pharmacological stimulation of the cholinergic antiin-31. flammatory pathway. J Exp Med 2002;195:781ñ788.Luyer MD, Greve JW, Hadfoune M, et al. Nutritional stimulation of cholecystokinin receptors 32. inhibits inflammation via the vagus nerve. J Exp Med 2005;202:1023ñ1029.Yu Z, Zhang W, Kone BC. Signal transducers and activators of transcription 3 (STAT3) inhibits 33. transcription of the inducible nitric oxide synthase gene by interacting with nuclear factor kap-paB. Biochem J 2002;367:97ñ105.Saeed RW, Varma S, Peng-Nemeroff T, et al. Cholinergic stimulation blocks endothelial cell 34. activation and leukocyte recruitment during inflammation. J Exp Med 2005;201:1113ñ1123.Kawashima K, Yoshikawa K, Fujii YX, et al. Expression and function of genes encoding cholin-35. ergic components in murine immune cells. Life Sci 2007;80:2314ñ2319.Giebelen IA, van Westerloo DJ, LaRosa GJ, et al. Stimulation of alpha 7 cholinergic receptors 36. inhibits lipopolysaccharide-induced neutrophil recruitment by a tumor necrosis factor alpha-independent mechanism. Shock 2007;27:443ñ447.

Page 118: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

5Pe

riphe

ral N

icot

inic

Ago

nist

s A

mel

iora

te P

osto

pera

tive

Ileus

117

Page 119: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

6

Page 120: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

6Cha

pter

6Central activation of the choli-

nergic anti-inflammatory path-

way shortens postoperative

ileus in mice

Frans O. The, Jan van der Vliet,

Wouter J. de Jonge, Roelof J. Bennink,

Ruud M. Buijs, Guy E. Boeckxstaens

submitted for publication

Page 121: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

120

AbstractBackground & Aims: Electrical stimulation of the vagus nerve reduces the intestinal

inflammation following mechanical handling thereby shortening postoperative ileus in

mice. Previous studies in a sepsis model showed that this cholinergic anti-inflammatory

pathway can be activated pharmacologically by central administration of semapimod,

a p38 MAPKinase inhibitor. Aim: To evaluate the effect of semapimod icv on intestinal

inflammation and postoperative ileus in mice. Methods: Mice underwent a laparotomy (L)

or intestinal manipulation (IM) 1h after pre-treatment with 1µg/kg semapimod or saline icv.

Drugs were administered through a cannula placed in the right lateral ventricle one week

prior to experiments. 24h after surgery, gastric emptying of a semi-liquid meal was measured

using scintigraphy and the degree of intestinal inflammation was assessed. Finally, brain

region activation was assessed using quantitative c-Fos immunohistochemistry. Values

are depicted as mean ± s.e.m. P<0.05 was considered statistically significant. Results: IM significantly delayed gastric emptying 24h after surgery in saline treated animals

(gastric retention at 80min (RT80) L=3±1%, vs. IM 19±4%, p<0.05, n=8) and induced

inflammation of the manipulated intestine (MPO-pos. cells/mm2: L= 48±7 vs. IM= 381±27,

p<0.05, n=8). Icv semapimod significantly reduced this inflammation and improved gastric

emptying (MPO-pos. cell/mm2: 227±28, p<0.05; RT80: 5±1%, p<0.05, n=8). Vagotomy

(VGX) enhanced IM induced inflammation and abolished the anti-inflammatory effect of

semapimod icv (MPO-pos.cell/mm2: VGX-saline 540±78 vs. saline, n=8, p<0.05 and vs.

VGX-semapimod 440±34, n=8, p=0.2). Semapimod but not saline induced a significant

increase in c-fos expression in the paraventricular nucleus, the nucleus of the solitary tract

and the dorsal motor nucleus of the vagus nerve. Conclusion: Our findings show that

icv semapimod reduces manipulation-induced intestinal inflammation and prevents POI

by central activation of the vagus nerve. In addition, we provide evidence suggesting that

semapimod activates the DMNV, possibly via activation of the paraventricular nucleus.

Page 122: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

6C

entra

l Act

ivat

ion

of th

e C

holin

ergi

c A

nti-i

nflam

mat

ory

Path

way

in P

osto

pera

tive

Ileus

121

TIntroductionThe vagus nerve plays a crucial role in the control of gastrointestinal function, including

secretion, visceral perception and motility. Recently, Tracey et al. provided strong evidence

indicating that the vagus nerve modulates the innate immune system1. They showed that

electrical stimulation of the vagus reduced TNF levels and prevented arterial hypotension

after endotoxin injection1. Similarly, we demonstrated that vagus nerve stimulation reduced

the inflammatory response to mechanical manipulation of the intestine during surgery and

thereby prevented surgery-induced delayed gastric emptying2. This anti-inflammatory effect

is mediated by acetylcholine interacting with nicotinic receptors located on macrophages,

leading to a reduction in macrophage activation and cytokine production3. This so-called

cholinergic anti-inflammatory pathway is suggested to represent an additional regulatory

system controlling the inflammatory response to a wide range of threats to the organism.

Inflammation is sensed by afferent nerve fibers and is subsequently sent to the brain stem

for integration4. After integration, the motor neurons of the vagus nerve are activated and

an integrated anti-inflammatory signal is sent back to the inflamed area4. The presence of

such a feedback loop/reflex and its anatomical connections are still hypothetical and still

need to be demonstrated. Nevertheless, this system may represent an interesting tool to

control inflammation in a number of disorders. In contrast to anti-inflammatory cytokines

and the hormonal control by corticosteroids (HPA axis), this neural system provides an

integrated response that is lightning fast and target specific. Obviously, it may provide new

therapeutic means to control or dampen inflammation, not only in case of sepsis or ileus, but

most likely also in other inflammatory diseases like rheumatoid arthritis and inflammatory

bowel diseases.

Semapimod, a tetravalent guanyl-hydrazone also known as CNI-1493, prevents

macrophage activation via inhibition of mitogen activated protein kinase signaling5. While

studying the effect of semapimod in cerebral ischemia, Meistrell et al. found that central

application of this drug could reduce systemic inflammation6. Further studies revealed

that semapimod, when infused intracerebroventricular (icv), is up to 100.000 times more

effective compared to intravenous administration (iv)7. In addition, electrophysiological

studies have shown enhanced activity of the vagus nerve after infusion of semapimod8.

Page 123: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

122

These findings strongly suggest that semapimod represents a pharmacological and central

activator of the cholinergic anti-inflammatory pathway.

Animal studies on the pathogenesis of postoperative ileus have shown that gentle small

bowel manipulation during abdominal surgery results in a distinct inflammation of the

muscularis propria9, 10. This local innate inflammatory response activates an adrenergic

inhibitory neural reflex leading to generalized hypomotility or ileus10. Reduction of the

inflammatory response by pre-treatment with intercellular adhesion molecule (ICAM)-1

inhibitory antibodies or antisense oligonucleotides, normalizes gastric emptying10, 11

further illustrating its crucial role in the pathogenesis of postoperative ileus. Previously, we

showed that both electrical stimulation of the vagus nerve2 and systemic administration of

selective nicotinic agonists12 had an anti-inflammatory effect on surgery-induced intestinal

inflammation, suggesting that activation of the cholinergic anti-inflammatory pathway

indeed may represent an interesting approach to treat intestinal inflammation. In the

present study, we evaluated whether pharmacological activation of the vagus nerve by

central application of semapimod also leads to reduced inflammation and prevention of

ileus. In addition, we performed c-fos immunohistochemical analysis of the brain stem to

illustrate the involvement of the motor nucleus of the vagus nerve.

Page 124: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

6C

entra

l Act

ivat

ion

of th

e C

holin

ergi

c A

nti-i

nflam

mat

ory

Path

way

in P

osto

pera

tive

Ileus

123

MethodsAnimalsFemale Balb/C mice (Harlan Nederland, Horst, The Netherlands), age 12 to 15 weeks,

were kept under environmentally controlled conditions (light on from 8:00 AM till 8:00 PM;

water and rodent nonpurified diet ad libitum; temperature 20°C-22°C; 55% humidity). All

experiments were performed with the approval of the Ethical Animal Research Committee

of the University of Amsterdam and according to their guidelines.

Study protocolsFirst, the efficacy of icv administered semapimod was evaluated in our mouse model of

postoperative ileus10. An icv cannula was placed in the left lateral ventricle of the brain 7 days

prior to surgery, as described below. Sixty min before the surgical procedure, animals were

treated with semapimod 1ug/kg icv or its vehicle (saline) in a volume of 5µl administered

in 10min, using an infusion (pump 22 multiple syringe pump, Harvard Apparatus, Holliston,

MA, USA). Twenty-four hrs after surgery, gastric emptying of a semi-liquid non-caloric test

meal was determined using a scintigraphic imaging technique 13. After completion, mice

were sacrificed by cervical dislocation and ileal segments (4-6 cm proximal of cecum) were

quickly excised for the assessment of intestinal inflammation.

In a different set of experiments, a subdiaphragmal bilateral vagotomy was performed

30min prior to infusion of semapimod or vehicle to determine vagus nerve involvement. To

identify the brain nuclei involved in the central activation of the cholinergic anti-inflammatory

pathway, c-fos expression was studied after icv treatment with semapimod vs. saline. A

swivel equipped infusion pump was used to administer the drugs, allowing the animals to

move freely in their usual environment. Swivel pumps were connected at 8 am in all animals

and infusion was started only after 4 hrs to minimize stress-induced brain activity. Three

hrs after icv administration of saline or semapimod, mice were transcardially perfused (1.6

mL/min) with 8 mL of a 0.9% NaCl solution, followed by 50 mL of 4% paraformaldehyde

in phosphate buffer (0.1 mol/L; pH 7.4). After perfusion, the brain, brainstem and proximal

spinal cord were carefully removed, postfixed overnight in the same fixative at 4°C, and

cryoprotected until further analysis in 30% sucrose solution containing 0.05% sodium azide

at 4°C.

Page 125: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

124

ICV cannula placementIn anesthetized animals, a cannula (23 G needle) was stereotaxically implanted into the left

lateral cerebral ventricle using the following coordinates from Bregma: 0.46mm posterior,

1.0 mm lateral and 2.2 mm ventral. Dental cement was used to secure the cannula to three

screws inserted into the skull.

Surgical procedureAnesthetized mice underwent a laparotomy (L) or a laparotomy followed by small intestinal

manipulation (IM) as described previously10. In short, a midline incision was made and

the peritoneal cavity was opened along the linea alba under sterile conditions. The small

intestine was carefully exteriorized from the distal duodenum until the cecum and gently

manipulated for 5 minutes using sterile moist cotton applicators. Contact or stretch

of stomach or colon was strictly avoided. After repositioning of the intestinal loops, the

abdomen was closed using a two-layer continuous suture (Mercilene Softsilk 6-0). Mice

recovered from surgery in a temperature controlled cage set at 32° C with free access

to water but not to food. Twenty-four hrs after surgery, gastric emptying was measured.

Thereafter, mice were anaesthetized and killed by cervical dislocation. The small intestine

was removed, flushed in ice-cold phosphate buffered saline (PBS), and snap frozen in

liquid nitrogen or fixed in ethanol for further analysis.

Subdiaphragmal vagotomyA midline incision was made and a retractor was placed. Under microscopic view, both

the left and right vagal nerve trunks were cut, distal from the diaphragm but proximal

to the division of the hepatic branch. During this procedure, the intraperitoneal organs

were protected and kept moist using sterile gausses drenched in NaCl. Any palpation or

manipulation of the small intestine was carefully avoided. The abdomen was closed using

a two-layer continuous suture (Mercilene Softsilk 6-0). Animals were kept in a temperature-

controlled cage at 32° C until drug infusion and surgery. Microscopic inspection and

postmortem evaluation of the stomach distention were utilized to determine a successful

vagotomy procedure.

Page 126: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

6C

entra

l Act

ivat

ion

of th

e C

holin

ergi

c A

nti-i

nflam

mat

ory

Path

way

in P

osto

pera

tive

Ileus

125

Measurement of gastric emptyingAs previously described, gastric emptying rate was determined after gavage of a semi-

liquid, non-caloric test meal (0.1ml of 3% methylcellulose solution containing 10 MBq of

99mTc-Albures11, 13. Mice were scanned using a gamma camera set at 140 keV13. The entire

abdominal region was scanned for 30 seconds, immediately and 80 minutes after gavage.

During the scanning period mice were conscious and manually restrained. The static images

obtained were analyzed using Hermes computer software (Hermes, Stockholm, Sweden).

Gastric retention was calculated by determining the percentage of activity present in the

gastric region of interest compared to the total abdominal region of interest11.

Quantification of intestinal muscle inflammation After sacrifice, the mesentery was removed from the intestine, which was cut open along

its mesenteric border. Fecal content was washed out in ice-cold PBS and fixed in 100%

ethanol for 10 minutes. Fixed preparations were kept in 70% ethanol at 4°C until further

analysis. Before final analysis segments were stretched 1.5 times to their original size and

pinned down on a glass-dish filled with 70% ethanol after which the mucosa was carefully

removed.

Myeloperoxidase (MPO) was stained using the method described in the specified section.

For quantification, the number of MPO-positive cells in five randomly chosen 1mm2 fields

was counted.

Quantification of brain regional C-fos expression4The number of C-fos positive nuclei were counted in the nucleus of the solitary tract (NTS),

the dorsal motor nucleus of the vagus nerve (DMNV) and the paraventricular nucleus

(PVN) in 4 to 8 section of each individual animal and divided by the number of sections.

These mean numbers of C-fos positive nuclei per animal were used for further statistical

analysis.

Page 127: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

126

Myeloperoxidase stainingFixed preparations were rehydrated by incubation in 50% ETOH and phosphate buffered

saline pH 7.4 for 5 minutes. To visualize myeloperoxidase(MPO)-positive cells preparations

were incubated for 10 minutes with 3-amino-9-ethyl carbazole (Sigma, St. Louis, MO) as a

substrate, dissolved in sodium acetate buffer (pH 5.0) to which 0.01% H2O2 was added10.

ImmunohystochemistryC-fos immunohistochemistry was performed according to Bonaz et al.14, with modifications.

After fixation, the brain was embedded in Tissue-Tek (Sakura Finetek Inc., Torrance, CA)

and 40mm transversal sections were cryostat-cut. Free-floating sections were washed with

Tris-buffered saline (TBS; pH 7.4) 3 times and incubated overnight at 4°C with the primary

polyclonal sheep antibody (0.3 µg/mL; Sigma Genosys, St. Louis, MO) in 0.25% gelatin

and 0.5% TritonX-100 in TBS. Next, sections were washed in TBS (3x) and incubated

with biotinylated anti-sheep antiserum (Vector Laboratories, Burlingame, CA) for 1.5 hrs at

room temperature. After washing in TBS (3x), sections were processed for avidin– biotin–

peroxidase (Vectorstain; Vector Laboratories), and peroxidase was visualized by using

diaminobenzidine in 0.02% nickel sulphate in TBS as the chromogen.

StatisticsStatistical analysis was performed using SPSS 12.02 software for Windows. The data were

non-parametrically distributed and therefore analyzed using the non-parametric Mann-

Whitney test. P<0.05 was considered statistically significant and results were depicted as

mean ± SEM.

Page 128: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

6C

entra

l Act

ivat

ion

of th

e C

holin

ergi

c A

nti-i

nflam

mat

ory

Path

way

in P

osto

pera

tive

Ileus

127

ResultsSemapimod administered icv ameliorates POI and diminishes manipulation-induced intestinal muscle inflammation. Manipulation of the small intestine during abdominal surgery (IM) initiated a significant

increase in gastric retention 24 hrs after the procedure when compared to mice undergoing

laparotomy (L) alone (gastric retention 80 min after gavage of test meal (GR80) IMsaline

19 ± 4% vs. Lsaline 3 ± 1%, n=8, p<0.001) (fig.1). The gastric stasis marking the extent

of postoperative ileus was accompanied by a marked myeloperoxidase(MPO)-positive

inflammatory cell influx in the manipulated segment. This local leukocyte recruitment was

not observed in L animals (MPO-positive cells/mm2 IMsaline 381 ± 27 cells/mm2 vs. Lsaline 48

± 7 cells/mm2, n=8, p=0.001) (fig.2). Treatment with semapimod 1µg/kg icv ameliorated the

IM-induced delay in gastric emptying (GR80 IMsemapomod 5 ± 1%, n=8, p= 0.02) (fig. 1). In

line with this observation, the number of MPO-positive cells in the intestinal muscle layer

also diminished significantly in the semapimod treated group compared to saline treated

control animals (IMsemapimod 227 ± 28 cells/mm2, n=8, p=0.003 vs. saline) (fig.2). In contrast,

semapimod did not alter gastric retention nor intestinal muscle inflammation compared to

saline in mice that underwent a L (GR80 Lsemapimod 5 ± 2%, n=8, p=0.4; MPO Lsemapimod 36 ±

9 cells/mm2, n=8, p=0.3) (fig.1 and 2).

saline semapimod saline semapimod

laparotomy intestinalmanipulation

0

10

20

30 p<0.001p=0.02

rela

tive

gast

ric c

onte

nts

(%) Figure 1 Effect of saline

(vehicle) or semapimod icv pre-treatment on gastric retention 24 hrs after laparotomy (L) or laparotomy followed by gentle intestinal manip-ulation (IM). Gastric re-tention was determined 80 min. after gavage of a semi-liquid test meal. Data are expressed as mean ± SEM (Mann-Whitney U test).

Page 129: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

128

The anti-inflammatory effect of semapimod is mediated through the vagus nerve.To assess the involvement of the vagus nerve, experiments were repeated in subdiaphragmal

vagotomized (VGX) animals. As gastric motility is strongly effected by VGX, gastric

emptying was not assessed. There was a significant inflammatory response 24hrs after

IM not seen 24hrs after L in mice subjected to VGX prior to abdominal surgey (IMvgx 540

± 78 cells/mm2 vs. Lvgx 52 ± 9 cells/mm2, n=8, p=0.004). This inflammatory response was

even severe when compared to the response observed in non-VGX animals undergoing

IM (IMVGX vs. IM, n=8, p=0.04) . In contrast, the anti-inflammatory effect in semapimod pre-

treated animals was absent after subdiaphragmal VGX (semapimod IMvgx 440 ± 34.3cells/

mm2 vs. saline IMvgx 404 ± 34, n=8, p=0.4) (fig. 2) illustrating that the anti-inflammatory

effect achieved with semapimod is mediated through vagus nerve signaling.

Semapimod induced c-fos expression in brain stem nuclei.To obtain more insight in the central mechanism through which semapimod activates the

cholinergic anti-inflammatory pathway, c-fos immunohistochemical analysis of the brain

was performed. Based on a previous report demonstrating enhanced activity of vagal

efferent nerve fibers upon semapimod administration8 we first assessed c-fos expression

in the dorsal motor nucleus of the vagus nerve (DMNV) and the nucleus of the solitary tract

(NTS). Quantitative c-fos analysis 3 hrs after infusion of semapimod showed a significant

increased number of c-fos positive neurons in both the NTS (saline 23 ± 3 vs. semapimod

38 ± 5, n=6, p=0.02) and the DMNV (saline 4 ± 0 vs. 8 ± 0, n=6, p=0.002) when compared

to saline (fig. 3b-c).

Vagal efferent control of pancreatic protein secretion is mediated through M1-receptor

stimulation in the paraventricular nucleus (PVN).15 Moreover, Pavlov et al. recently found

that semapimod competitively binds to M1-muscarinic receptors.16 These observations led

to our hypothesis that semapimod triggers neurons in the PVN projecting to the dorsal

motor complex of the vagus nerve (DVC) hereby activating the cholinergic anti-inflammatory

pathway. Therefore we assessed c-fos activation in the PVN 3hrs after icv injection of

semapimod and found that c-fos expression was significantly increased in the PVN after

semapimod compared to saline infusion (19 ± 5 vs. 70 ± 23, n=6, p=0.03)(fig. 3a).

Page 130: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

6C

entra

l Act

ivat

ion

of th

e C

holin

ergi

c A

nti-i

nflam

mat

ory

Path

way

in P

osto

pera

tive

Ileus

129

saline VGX saline

semapimod VGX semapimod

saline VGX saline

semapimod VGX semapimod

0

100

200

300

400

500

600

700M

PO-p

os. c

ells

/mm

2

p=0.001

p=0.003

p=0.04

lanitsetniymotorapalmanipulation

p=0.004

Figure 2 Effect of saline (vehicle) or semapimod icv pre-treatment on manipulation-induced inflam-matory cell recruitment to the muscularis propria. Quantative analysis of the number of MPO-positive cells 24 hrs after laparotomy (L) or laparotomy followed by gentle intestinal manipulation (IM). Mice that underwent a subdiaphragmal vagotomy prior to treatment are marked by VGX. Data are ex-pressed as mean ± SEM (Mann-Whitney U test).

Page 131: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

130

DMNV

saline DMNV semapimod DMNV0

2

4

6

8

10 p=0.002

saline PVN semapimod PVN0

20

40

60

80

100

p=0.03PVN

NTS

saline NTS semapimod NTS0

20

40

60

80

100 p=0.04

A

B

num

ber o

f c-F

OS

pos.

neu

rons

num

ber o

f c-F

OS

pos.

neu

rons

num

ber o

f c-F

OS

pos.

neu

rons

C

Figure 3 C-fos expression in in the A) PVN, B) DMNV and C) NTS 3 hrs after icv saline or semapimod treatment. Data are expressed as mean ± SEM (Mann-Whitney U test).

Page 132: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

6C

entra

l Act

ivat

ion

of th

e C

holin

ergi

c A

nti-i

nflam

mat

ory

Path

way

in P

osto

pera

tive

Ileus

131

DiscussionManipulation-induced inflammation of the intestine, a process orchestrated by innate

immune cells like mast cells and macrophages10, 17-23, is now generally believed to play

an imperative role in the pathophysiology of prolonged postoperative ileus. Recently, we

showed that vagus nerve stimulation reduces this inflammation and thereby shortens

postoperative ileus. Semapimod, a p38 MAPKinase inhibitor, has been shown to be a

central pharmacological activator of this so-called cholinergic anti-inflammatory pathway7, 8.

In the present study we showed that 1. icv semapimod reduces the inflammatory response

to intestinal manipulation and restores gastric emptying, 2. the anti-inflammatory effect

of semapimod is abolished by vagotomy, 3. icv semapimod leads to increased c-fos

expression in the PVN and DMNV. These findings confirm that central administration of

semapimod activates the cholinergic anti-inflammatory pathway leading to a reduction of

the manipulation-induced intestinal inflammation and restoration of gastric emptying.

Macrophages, present as a network between the circular and longitudinal muscle layers of

the intestine19, have been shown to play an important role in the pathogenesis of sustained

postoperative ileus in rodents20, 21. These phagocytes lay in close proximity of the myenteric

plexus and carry nicotinic acetylcholine receptors enabling neuro-immune interaction2.

Recently we demonstrated that the manipulation-induced inflammation can be diminished by

electrical stimulation of the vagus nerve in our experimental mouse model for postoperative

ileus2 A similar anti-inflammatory effect of vagus nerve stimulation has been demonstrated

in sepsis, ischemia-reperfusion, IBD and pancreatitis1, 24-26, and is currently referred to as

the cholinergic anti-inflammatory pathway. Stimulation of the efferent vagus nerve results

in reduction of pro-inflammatory cytokine release, i.e. TNF, IL1β and IL6, hereby improving

outcome in experimental septic-shock1. The peripheral mechanism of this cholinergic

anti-inflammatory response is mediated through alpha-7 nicotinergic receptors expressed

on macrophages3. Activation of these receptors results in JAK2/STAT3 signaling2 and

inhibition of NF-κB signal transduction27. Indeed, systemic application of selective alpha-7

nicotinergic agonists mimics the effect of vagus nerve stimulation in experimental models

for inflammatory bowel disease, pancreatitis and postoperative ileus.12, 24, 25

Page 133: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

132

Semapimod, a tetravalent guanylhydrazone also known as CNI-1493, has been suggested

a central, pharmacological activator of the cholinergic anti-inflammatory pathway.7,

8 Here we found that semapimod indeed suppresses inflammation in our mouse model

for postoperative ileus. Application of 1ug/kg semapimod administered icv diminished

manipulation induced inflammation and normalized gastric emptying. This effect was

abolished by subdiaphragmal vagotomy indicating central activation of the vagus nerve.

Pavlov et al. have demonstrated high affinity of semapimod for M1 muscarinic receptors16.

Moreover, they showed activation of the cholinergic anti-inflammatory pathway by central

administration of muscarinic agonists such as mucarine and the M1 selective agonist MCN-

A-343 with reduction of TNF release in an endotoxemia model16. However, the exact brain

areas involved remain to be clarified. In line with our functional data and the transient

increase of activity on vagal efferent recordings upon icv semapimod8, we observed an

increase in the number of c-fos positive neurons in the DMNV after semapimod but not

after saline. Accepting that semapimod interacts with central M1 receptors (Pavlov et al.)16,

direct activation of the DMNV is rather unlikely as this brain nucleus lacks M1 receptors28.

Previously, M1-receptor mediated activation of vagal output controlling pancreatic protein

secretion was shown to be mediated through stimulation in the paraventricular nucleus

(PVN).15 In line with this, we demonstrated c-fos activation in the PVN after icv administration

of semapimod. This finding and the knowledge that the PVN is interconnected with the

DMNV29, suggest that activation of the cholinergic anti-inflammatory pathway by semapimod

is indirect via interaction with M1 receptors in the PVN. Further studies however are required

to further confirm this hypothesis. In addition to increased c-fos expression in the DMNV,

we also observed enhanced c-Fos expression in the NTS following i.c.v. administration

of semapimod. This is in line with electrophysiological findings by Zhang et al.30 These

authors indeed demonstrated that although NTS neurons are predominantly inhibited, a

minority of NTS neurons was activated by electrical stimulation of PVN neurons.

Interestingly, we also demonstrated an increase in the inflammatory response to intestinal

manipulation in vagotomized animals. Similar findings have been reported in other models

of inflammation8 and of sepsis1. For example, vagotomy increased the mortality rate in

animals subjected to hemorrhagic shock, associated with an increase in TNF levels31.

Similarly, the degree of DSS colitis25 and pancreatitis24 was significantly augmented after

Page 134: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

6C

entra

l Act

ivat

ion

of th

e C

holin

ergi

c A

nti-i

nflam

mat

ory

Path

way

in P

osto

pera

tive

Ileus

133

vagotomy. Together with our findings, these data would suggest endogenous activation

of the cholinergic anti-inflammatory pathway by the ongoing peripheral inflammatory

response, and would fit with the hypothesis that the vagus nerve exerts an important role

in modulating the innate immune system.

In conclusion, we showed that icv administration of semapimod reduces intestinal

inflammation and postoperative ileus induced by abdominal surgery via activation of the

cholinergic anti-inflammatory pathway. In addition, we provided indirect evidence that

semapimod activates the DMNV via activation of the PVN. These findings further demonstrate

the anti-inflammatory properties of the cholinergic anti-inflammatory pathway.

Page 135: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

134

Reference ListBorovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI, Watkins LR, Wang H, Abumrad N, 1. Eaton JW, Tracey KJ. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 2000;405:458-462.de Jonge WJ, van der Zanden EP, The FO, Bijlsma MF, van Westerloo DJ, Bennink RJ, 2. Berthoud HR, Uematsu S, Akira S, van den Wijngaard RM, Boeckxstaens GE. Stimulation of the vagus nerve attenuates macrophage activation by activating the Jak2-STAT3 signaling pathway. Nat.Immunol. 2005;6:844-851.Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S, Li JH, Wang H, Yang H, Ulloa L, 3. Al Abed Y, Czura CJ, Tracey KJ. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature 2003;421:384-388.Tracey KJ. Physiology and immunology of the cholinergic antiinflammatory pathway. J Clin 4. Invest 2007;117:289-96.Lowenberg M, Verhaar A, van den BB, ten Kate F, van Deventer S, Peppelenbosch M, Hom-5. mes D. Specific inhibition of c-Raf activity by semapimod induces clinical remission in severe Crohn’s disease. J.Immunol. 2005;175:2293-2300.Meistrell ME, III, Botchkina GI, Wang H, Di Santo E, Cockroft KM, Bloom O, Vishnubhakat JM, 6. Ghezzi P, Tracey KJ. Tumor necrosis factor is a brain damaging cytokine in cerebral ischemia. Shock 1997;8:341-348.Bernik TR, Friedman SG, Ochani M, DiRaimo R, Ulloa L, Yang H, Sudan S, Czura CJ, Ivanova 7. SM, Tracey KJ. Pharmacological stimulation of the cholinergic antiinflammatory pathway. J.Exp.Med. 2002;195:781-788.Borovikova LV, Ivanova S, Nardi D, Zhang M, Yang H, Ombrellino M, Tracey KJ. Role of vagus 8. nerve signaling in CNI-1493-mediated suppression of acute inflammation. Auton.Neurosci. 2000;85:141-147.Kalff JC, Carlos TM, Schraut WH, Billiar TR, Simmons RL, Bauer AJ. Surgically induced leuko-9. cytic infiltrates within the rat intestinal muscularis mediate postoperative ileus. Gastroenterol-ogy 1999;117:378-387.de Jonge WJ, van den Wijngaard RM, The FO, ter Beek ML, Bennink RJ, Tytgat GN, Buijs 10. RM, Reitsma PH, van Deventer SJ, Boeckxstaens GE. Postoperative ileus is maintained by intestinal immune infiltrates that activate inhibitory neural pathways in mice. Gastroenterology 2003;125:1137-1147.The FO, de Jonge WJ, Bennink RJ, van den Wijngaard RM, Boeckxstaens GE. The ICAM-1 11. antisense oligonucleotide ISIS-3082 prevents the development of postoperative ileus in mice. Br.J.Pharmacol. 2005.The FO, Boeckxstaens GE, Snoek SA, Cash JL, Bennink R, Larosa GJ, van den Wijngaard 12. RM, Greaves DR, de Jonge WJ. Activation of the cholinergic anti-inflammatory pathway ame-liorates postoperative ileus in mice. Gastroenterology 2007;133:1219-28.Bennink RJ, de Jonge WJ, Symonds EL, van den Wijngaard RM, Spijkerboer AL, Benninga 13. MA, Boeckxstaens GE. Validation of gastric-emptying scintigraphy of solids and liquids in mice using dedicated animal pinhole scintigraphy. J.Nucl.Med. 2003;44:1099-1104.Bonaz B, Plourde V, Tache Y. Abdominal surgery induces Fos immunoreactivity in the rat brain. 14. J.Comp Neurol. 1994;349:212-222.Li Y, Wu X, Zhu J, Yan J, Owyang C. Hypothalamic regulation of pancreatic secretion is medi-15. ated by central cholinergic pathways in the rat. J.Physiol 2003;552:571-587.Pavlov VA, Ochani M, Gallowitsch-Puerta M, Ochani K, Huston JM, Czura CJ, Al Abed Y, 16. Tracey KJ. Central muscarinic cholinergic regulation of the systemic inflammatory response during endotoxemia. Proc.Natl.Acad.Sci.U.S.A 2006;103:5219-5223.Wehner S, Schwarz NT, Hundsdoerfer R, Hierholzer C, Tweardy DJ, Billiar TR, Bauer AJ, Kalff 17. JC. Induction of IL-6 within the rodent intestinal muscularis after intestinal surgical stress. Sur-gery 2005;137:436-446.

Page 136: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

6C

entra

l Act

ivat

ion

of th

e C

holin

ergi

c A

nti-i

nflam

mat

ory

Path

way

in P

osto

pera

tive

Ileus

135

Kalff JC, Schraut WH, Billiar TR, Simmons RL, Bauer AJ. Role of inducible nitric oxide syn-18. thase in postoperative intestinal smooth muscle dysfunction in rodents. Gastroenterology 2000;118:316-327.Mikkelsen HB. Macrophages in the external muscle layers of mammalian intestines. Histol.19. Histopathol. 1995;10:719-736.Wehner S, Behrendt FF, Lyutenski BN, Lysson M, Bauer AJ, Hirner A, Kalff JC. Inhibition of 20. macrophage function prevents intestinal inflammation and postoperative ileus in rodents. Gut 2006.Kalff JC, Schraut WH, Simmons RL, Bauer AJ. Surgical manipulation of the gut elicits an intes-21. tinal muscularis inflammatory response resulting in postsurgical ileus. Ann.Surg. 1998;228:652-663.Kalff JC, Turler A, Schwarz NT, Schraut WH, Lee KK, Tweardy DJ, Billiar TR, Simmons RL, 22. Bauer AJ. Intra-abdominal activation of a local inflammatory response within the human mus-cularis externa during laparotomy. Ann.Surg. 2003;237:301-315.de Jonge WJ, The FO, van der CD, Bennink RJ, Reitsma PH, van Deventer SJ, van den 23. Wijngaard RM, Boeckxstaens GE. Mast cell degranulation during abdominal surgery initiates postoperative ileus in mice. Gastroenterology 2004;127:535-545.van Westerloo DJ, Giebelen IA, Florquin S, Bruno MJ, Larosa GJ, Ulloa L, Tracey KJ, van der 24. PT. The vagus nerve and nicotinic receptors modulate experimental pancreatitis severity in mice. Gastroenterology 2006;130:1822-1830.Ghia JE, Blennerhassett P, Kumar-Ondiveeran H, Verdu EF, Collins SM. The vagus nerve: a 25. tonic inhibitory influence associated with inflammatory bowel disease in a murine model. Gas-troenterology 2006;131:1122-30.Bernik TR, Friedman SG, Ochani M, DiRaimo R, Susarla S, Czura CJ, Tracey KJ. Cholinergic 26. antiinflammatory pathway inhibition of tumor necrosis factor during ischemia reperfusion. J.Vasc.Surg. 2002;36:1231-1236.Wang H, Liao H, Ochani M, Justiniani M, Lin X, Yang L, Al Abed Y, Wang H, Metz C, Miller EJ, 27. Tracey KJ, Ulloa L. Cholinergic agonists inhibit HMGB1 release and improve survival in experi-mental sepsis. Nat.Med. 2004;10:1216-1221.Hoover DB, Hancock JC, DePorter TE. Effect of vagotomy on cholinergic parameters in nuclei 28. of rat medulla oblongata. Brain Res Bull 1985;15:5-11.Rogers RC, Kita H, Butcher LL, Novin D. Afferent projections to the dorsal motor nucleus of the 29. vagus. Brain Res Bull 1980;5:365-73.Zhang X, Fogel R, Renehan WE. Stimulation of the paraventricular nucleus modulates the 30. activity of gut-sensitive neurons in the vagal complex. Am.J.Physiol 1999;277:G79-G90.Guarini S, Cainazzo MM, Giuliani D, Mioni C, Altavilla D, Marini H, Bigiani A, Ghiaroni V, Pas-31. saniti M, Leone S, Bazzani C, Caputi AP, Squadrito F, Bertolini A. Adrenocorticotropin reverses hemorrhagic shock in anesthetized rats through the rapid activation of a vagal anti-inflammato-ry pathway. Cardiovasc Res 2004;63:357-65.

Page 137: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

7

Page 138: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

7Cha

pter

7Mast Cell Degranulation Du-

ring Abdominal Surgery

Initiates Postoperative Ileus in

Mice

Wouter J. De Jonge, Frans O. The,

Dennis van der Coelen, Roelof J. Bennink, Pieter H. Reitsma,

Sander J. van Deventer, René M. van den Wijngaard

Guy E. Boeckxstaens

Gastroenterology 2004; 127: 535-545

Page 139: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

138

AbstractBackground & Aims: Inflammation of the intestinal muscularis following manipulation

during surgery plays a crucial role in the pathogenesis of postoperative ileus. Here, we

evaluate the role of mast cell activation in the recruitment of infiltrates in a murine model.

Methods: Twenty-four hours after control laparotomy or intestinal manipulation, gastric

emptying was determined. Mast cell degranulation was determined by measurement of mast

cell protease-I in peritoneal fluid. Intestinal inflammation was assessed by determination of

tissue myeloperoxidase activity and histochemical staining. Results: Intestinal manipulation

elicited a significant increase in mast cell protease-I levels in peritoneal fluid and resulted

in recruitment of inflammatory infiltrates to the intestinal muscularis. This infiltrate was

associated with a delay in gastric emptying 24 hours after surgery. Pretreatment with

mast cell stabilizers ketotifen (1 mg/kg, PO) or doxantrazole (5 mg/kg, IP) prevented both

manipulation-induced inflammation and gastroparesis. Reciprocally, in vivo exposure of

an ileal loop to the mast cell secretagogue compound 48/80 (0.2 mg/mL for 1 minute)

induced muscular inflammation and delayed gastric emptying. The manipulation-induced

inflammation was dependent on the presence of mast cells because intestinal manipulation in

mast cell-deficient Kit/Kitv mice did not elicit significant leukocyte recruitment. Reconstitution

of Kit/Kitv mice with cultured bone marrow-derived mast cells from congenic wild types

restored the manipulation-induced inflammation. Conclusions: Our results show that

degranulation of connective tissue mast cells is a key event for the establishment of the

intestinal infiltrate that mediates postoperative ileus following abdominal surgery.

Page 140: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

7M

ast c

ell D

egra

nula

tion

Initi

ates

in P

osto

pera

tive

Ileus

139

PBackgroundPostoperative ileus (POI) is characterized by dysmotility of the gastrointestinal tract that

occurs after essentially every abdominal procedure.1,2 Recent evidence indicates that

postoperative ileus following bowel manipulation is a biphasic process. An acute phase of

generalized enteric hypomotility is due to activation of inhibitory neural reflexes,3,4 which

is dependent on the release of α-calcitonin gene-related peptide (CGRP)5–7 and central

corticotropin-releasing factor.8 A subsequent prolonged phase is mediated by inflammation

of the intestinal muscularis externa that is induced by mechanical manipulation of the

gut.9–12 The muscular inflammation following bowel manipulation results in postoperative

motility changes of the manipulated small intestinal segment, i.e., impaired contractility and

delayed transit.10–12 However, the duration of POI is not determined by hampered peristalsis

of the small intestine only but rather by hypomotility of the entire gastrointestinal tract. In

this light, we recently showed that the inflammatory infiltrates in the small intestine not

only impair the neuromuscular function of the manipulated small intestine but also lead to

impaired gastric emptying.9 This gastroparesis resulted from the activation of an inhibitory

adrenergic neural pathway triggered by the intestinal infiltrates, explaining the generalized

nature of POI. Inflammatory infiltrates recruited to the bowel wall after manipulation are thus

crucial in the pathogenesis of POI. The mechanism as to how mechanical manipulation of

the intestine induces inflammation has been shown to involve the activation of a resident

macrophage network in the intestinal muscularis.10–12 The exact nature of the trigger behind

activation of these macrophages, however, remains unknown.

In this respect, intestinal manipulation has previously been described to initiate extensive

mast cell activation and degranulation.13 The murine intestine contains mast cells of the

mucosal (MMC) and connective tissue subtype (CTMC) that have distinct expression

of proteases.14 Intestinal mast cells contain numerous substances released upon

degranulation that are potent proinflammatory mediators such as tumor necrosis factor

(TNF) α,15 macrophage inflammatory protein-2 (MIP-2),15 and interleukin (IL)-8.16 Mast cells

have been shown to mediate granulocyte infiltration in a number of inflammatory conditions

involving delayed-type hypersensitivity reactions15 or in other diseases, such as bullous

pemphigoid,17 intestinal ischemia,18 and asthma.19 We therefore hypothesized that mast

cell degranulation may also initiate manipulation-induced intestinal inflammation in POI.

Page 141: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

140

In the current study, we provide evidence that mast cells degranulate upon intestinal

manipulation and that mast cell degranulation initiates the muscularic inflammation that

mediates POI. By employing 2 mast cell stabilizers, doxantrazole, as a nonselective

stabilizer that acts on MMC as well as CTMC, and ketotifen, which stabilizes only CTMC,20

we demonstrate that CTMC are primarily involved in this process. Hence, these findings set

the stage for the clinical use of mast cell stabilizers to shorten POI.

Page 142: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

7M

ast c

ell D

egra

nula

tion

Initi

ates

in P

osto

pera

tive

Ileus

141

Materials and MethodsLaboratory AnimalsMice (female BalB/C, Harlan Nederland, Horst, The Netherlands) were kept under

environmentally controlled conditions (light on from 8:00 AM to 8:00 PM; water and rodent

nonpurified diet ad libitum; 20°C–22°C, 55% humidity). Mast cell-deficient WBB6F1-W/WV

(Kit/Kitv) and the congenic mast cell-sufficient W/+ (Kit/WT) littermates were purchased

from The Jackson Laboratory (Bar Harbor, ME). Mice were maintained at the animal facility

of the Academic Medical Center in Amsterdam and were used at 15–20 weeks of age.

Animal experiments were performed in accordance with the guidelines of the Ethical Animal

Research Committee of the University of Amsterdam.

Surgical Procedures: Abdominal SurgeryWith Intestinal Manipulation Mice were anesthetized by an intraperitoneal (IP) injection of a

mixture of fentanyl citrate/fluanisone (Hypnorm; Janssen, Beerse, Belgium) and midazolam

(Dormicum; Roche, Mijdrecht, The Netherlands). Surgery was performed under sterile

conditions. Mice (8–12 per treatment group) underwent control surgery of only laparotomy

or laparotomy followed by intestinal manipulation. The surgery was performed as follows:

A midline abdominal incision was made, and the peritoneum was opened over the linea

alba. The small bowel was carefully externalized, layered on a sterile moist gauze pad,

and manipulated from the distal duodenum to the cecum for 5 minutes, using sterile moist

cotton applicators. Contact or stretch on stomach or colon was strictly avoided. After the

surgical procedure, the abdomen was closed by a continuous 2-layer suture (Mersilene, 6-0

silk). After closure, mice were allowed to recover for 4 hours in a heated (32°C) recovery

cage with free access to drinking water but not food. At 4 hours postoperatively, mice were

completely recovered from anesthesia. At 24 hours after surgery, the gastric emptying rate

was measured using gastric scintigraphy. Thereafter, mice were anesthetized and killed by

cervical dislocation, and the small intestine was removed, flushed in ice-cold saline, and

snap frozen in liquid nitrogen or fixed in ice-cold ethanol for further analysis.

Page 143: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

142

Study Protocols The effect of doxantrazole or ketotifen treatment on postoperative intestinal inflammation

and gastric emptying. One group of mice received the mast cell stabilizer doxantrazole (5

mg/kg in 5% NaHCO3 , pH 7.4; a kind gift of Agne`s Francois, Institut Gustave Roussy,

Villejuif, France) or its vehicle, via IP injection once daily for 3 days.21 Alternatively,

ketotifen (1 mg per kg; Sigma Chemical Co., St. Louis, MO) in 0.5% methylcellulose

solution in water was administered by oral gavage once daily for 5 consecutive days.22

Ketotifen controls received only 0.5% methylcellulose in water. Mice underwent abdominal

surgery with intestinal manipulation 1 hour after the final treatment. Twenty-four hours after

surgery, gastric emptying was measured. Thereafter, the mice were killed, and the intestine

was isolated. Intestinal tissue was cut open along the mesenterial border, washed in ice-

cold saline, blotted dry, and frozen in liquid nitrogen for determination of MPO activity.

Alternatively, tissue was fixed in ice-cold 4% paraformaldehyde or ethanol and processed

for histologic analyses.

The effect of in vivo mast cell degranulation on intestinal inflammation and gastric emptying.

Local mast cell degranulation in the ileum was evoked as follows: A midline laparotomy

was performed, and 6 cm of ileum proximal to the cecum was carefully externalized and

placed in a sterile aluminum cup without touching or stretching other parts of the GI tract.

Of these 6 cm of ileum, the proximal 5-cm segment was incubated in either an 0.2 mg/mL

solution of compound 48/80 (C48/80, Sigma Co.) or vehicle (saline) for 1 minute at 37°C.

Leakage of C48/80 solution into the peritoneal cavity was strictly avoided. Segments were

incubated for a short period of only 1 minute to avoid potential systemic uptake of C48/80.

After incubation, the C48/80 solution was removed, and the ileal segment was washed 3

times with 0.9% NaCl, kept prewarmed at 37°C. After closure, the animals were allowed

to recover for 4 hours in a heated recovery cage. No mortality was observed following this

treatment.

The effect of intestinal manipulation in Kit/Kitv mice and mast cell reconstituted Kit/Kitv

mice. Kit/Kitv mice are completely devoid of mast cells in their gastrointestinal tract or

other anatomical sites.23 Kit/Kitv mice were reconstituted by the injection of bone marrow-

derived cultured mast cells into the peritoneal cavity, as described.24 In brief, femoral bone

marrow cells from Kit/WT control mice were maintained in vitro for 4 weeks in RPMI 1640

Page 144: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

7M

ast c

ell D

egra

nula

tion

Initi

ates

in P

osto

pera

tive

Ileus

143

complete medium (Life Technologies Inc., Grand Island, NY) supplemented with 10% fetal

calf serum, in the presence of stem cell factor (50 ng/mL; Pepro Tech, Rocky Hill, NJ) and

interleukin-3 (1 ng/mL; Pepro Tech, Rocky Hill, NJ). During culture, medium was refreshed

once weekly. After this culture period, mast cells represented more than 95% of the total

cells as determined by Toluidine blue staining on cytospin preparations. Subsequently, mast

cells were harvested, and 2 x 106 cells in 100 µL PBS were injected in Kit/Kitv mice. PBS

alone (100 µL IP) was injected as a negative control. This procedure reconstitutes the mast

cell population without systemic effects. 24 To confirm mast cell reconstitution, we stained

peritoneal cells obtained by lavage as well as intestinal, mesenteric, and gastric tissue

sections with Toluidine blue and Giemsa. Mice were used 10 weeks after adoptive transfer

of mast cells. Twenty-four hours after intestinal manipulation, intestinal and gastric tissue

was obtained and analyzed by Toluidine blue staining. Tissue was snap frozen in liquid

nitrogen and stored at -80°C for determination of MPO enzyme activity. Peritoneal cells

were harvested by lavage with 5 mL 0.9% NaCl, and the cell suspension was centrifuged at

400g for 5 minutes at 4°C. MMCP-1 levels were measured in blood plasma and peritoneal

lavage fluid by sandwich ELISA according to the manufacturer’s instructions (Moredun

Scientific, Edinburgh, Scotland).

Measurement of Gastric Emptying and TransitGastric emptying was determined as described previously.25 Gastric emptying rate

was determined after offering a caloric solid test meal (100 mg egg yolk containing 10

MBequerel(MBq) of 99mTc-Albures) that as consumed within 1 minute.26 Immediately after

complete consumption, mice were scanned using a gamma camera set at 140 keV. The

entire abdominal region was scanned for 30 seconds at 16-minute intervals for 112 minutes.

During the 30-second scanning period, mice were conscious and manually restrained.

The static images obtained were analyzed using Hermes computer software (Hermes,

Stockholm, Sweden). Gastric emptying was measured by determining the percentage of

activity present in the gastric region of interest, compared with the total abdominal region of

interest, for each image. Subsequently, the gastric half-emptying time (t1/2) was determined

for each individual mouse using DataFit software (version 6.1, Oakdale Engineering,

Oakdale, PA). A modified power exponential function y(t) = 1 - (1 - ekt)b was used, where

y(t) is the fractional meal retention at time t, k is the gastric emptying rate per minute, and

b is the extrapolated y-intercept from the terminal portion of the curve.

Page 145: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

144

Quantification of Leukocyte Accumulation at the Intestinal MuscularisMyeloperoxidase (MPO) activity in full-thickness ileal segments was assayed as a measure

of leukocyte infiltration as described elsewhere.9 Tissue was blotted dry, weighed, and

homogenized in a 20 times volume of a 20 mmol/L potassium phosphate buffer, pH

7.4. The suspension was centrifuged (8000g for 20 minutes at 4°C), and the pellet was

taken up in 1 mL of a 50 mmol/L potassium phosphate buffer, pH 6.0, containing 0.5% of

hexadecyltrimethylammoniumbromide (HETAB) and 10 mmol/L ethylenediaminetetraacetic

acid (EDTA). Fifty microliters of the appropriate dilutions of the tissue homogenate was

added to 445 µL of assay mixture, containing 0.2 mg/mL tetramethylbenzidine in 50 mg

potassium phosphate buffer, pH 6.0, 0.5% HETAB, and 10 mmol/L EDTA. The reaction

was started by adding 5 µL of a 30 mmol/L H2O2 to the assay mixture, and the mixture was

incubated for 3 minutes at 37°C. After 3 minutes, 30 µL of a 300 µg/mL catalase solution

was added to each tube, and tubes were placed on ice for 3 minutes. The reaction was

ended by adding 2 mL of 0.2 mol/L glacial acetic acid. Absorbance was read at 655 nm.

A standard reference curve was established using purified MPO (Sigma Co.). One unit of

MPO activity was defined as the quantity of MPO activity required to convert 1 µmol of H2O2

to H2O per minute at 25°C, using purified MPO activity as a standard (Sigma, St Louis,

MO). MPO content was expressed as units of MPO activity per milligram of tissue.

Whole Mount PreparationWhole mounts of ileal muscularis were prepared as previously described.9,11 In short,

ileal segments (2–6 cm distal from the cecum) were quickly excised, and mesentery was

removed. Ileal segments were cut open along the mesentery border, fecal content was

washed out in ice-cold PBS, and segments were pinned flat in a glass-dish filled with

preoxygenated Krebs-Ringer solution, pH 7.4. Mucosa was removed, and the remaining

full-thickness sheet of muscularis externa was fixed for 10 minutes in 100% ethanol.

Muscularis preparations were kept on 70% ethanol at 4°C until analysis.

Page 146: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

7M

ast c

ell D

egra

nula

tion

Initi

ates

in P

osto

pera

tive

Ileus

145

Statistical AnalysisThe data are expressed as mean ± SEM and were analyzed using the nonparametric

Mann–Whitney U test or 1-way ANOVA where indicated. A P value less than 0.05 was

considered significant.

Page 147: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

146

ResultsIntestinal Manipulation Triggers Intestinal Mast Cell DegranulationWe determined whether the gentle intestinal manipulation induces mast cell degranulation

in our experimental model of POI. To this end, the level of the mast cell-specific soluble

chymase murine mast cell proteinase-1 (mMCP-1) in the serum and peritoneal lavage fluid

was measured 20 minutes after intestinal manipulation. Intestinal manipulation led to a

significant increase in peritoneal mMCP-1, compared with a control laparotomy (Table 1).

Serum mMCP-1 levels were not significantly altered (not shown). Pretreatment with mast

cell stabilizing agents ketotifen or doxantrazole effectively prevented mast cell degranulation

during intestinal manipulation in that the increase in peritoneal mMCP-1 levels after

intestinal manipulation was not observed following intestinal manipulation with ketotifen or

doxantrazole pretreatment (Table 1).

Table 1. mMCP-1 levels in peritoneal lavage fluid (n=4-5),3 h PO det limit 1.25 ng/mL

LIMLLIMIM

treatment--shamVS 5VshamVS 5V

<1.34.1±0.5<1.3

3.5±0.9

<1.33.7±0.7

mMCP1 (ng/mL)surgery

Mast Cell Stabilization Prevents Intestinal Inflammation and the Development of POI Following Abdominal SurgeryWe aimed to investigate whether pretreatment with a mast cell stabilizer prevented

postoperative gastroparesis induced by intestinal manipulation. We measured gastric

emptying 24 hours after surgery in mice pretreated with either doxantrazole (5 mg/kg,

IP for 3 days once daily) or ketotifen (1 mg/kg, PO for 5 days once daily). As shown in

Figure 1, neither of the vehicles used for ketotifen or doxantrazole administration affected

basal gastric emptying (Figure 1A and B). However, intestinal manipulation significantly

increased halfemptying time and gastric retention of the test meal compared with control

mice treated with vehicle saline (Figure 1A) or 5% NaHCO3 (Figure 1B).

Page 148: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

7M

ast c

ell D

egra

nula

tion

Initi

ates

in P

osto

pera

tive

Ileus

147

rela

tive

gast

ric c

onte

nt (%

)

20

60

100

20 60 100time (min)

L+ vehicle

IM + vehicleIM + ketotifen

t (min)

30.5 ± 4.2 31.6 ± 2.447.5 ± 6.9 *30.3 ± 8.3

-+-+

pretreatment surgery

25.3 ± 4.726.5 ± 2.641.1 ± 5.2 * 27.4 ± 7.1

Ret60min (%)

28.8 ± 4.933.5 ± 10.759.1 ± 7.5 *43.3 ± 7.5

LLIMIM

-+-+

24.6 ± 5.633.3 ± 4.043.1 ± 5.2*39.0 ± 7.1*

ketotifen

doxantrazole

20 60 100

20

60

100

time (min)

L+ vehicle

IM + vehicleIM + doxantrazole

LLIMIM

A B

Figure 1. Delayed gastric emptying after intestinal manipulation is prevented by pretreatment with mast cell stabilizers ketotifen or doxantrazole. (A) Gastric emptying curves determined by scinti-graphic imaging of the abdomen after oral administration of solid caloric meal at 24 hours after intestinal manipulation (IM + vehicle; solid squares), or IM after pretreatment with ketotifen (open squares), compared with laparotomy (L + vehicle; gray circles). Values shown are percentages of gastric content compared with the total abdominal region. (B) Treatment with doxantrazole prevents gastroparesis 24 hours following bowel surgery (IM + doxantrazole), compared with vehicle treatment (IM + vehicle). Lower panel: corresponding deduced half-emptying time (t1/2) as well as the retention after 60 minutes. Ret60min is significantly increased after IM, irrespective of vehicle used, compared with L. Pretreatment with either ketotifen or doxantrazole restores both t1/2 to normal. Note that Ret60min is restored to normal only following ketotifen treatment. Treatment with ketotifen or doxan-trazole did not alter basal emptying after L. Values are averages ± SEM of 5–10 mice per treatment group. Asterisks indicate significant differences with respective L + vehicle group at P < 0.05.

In contrast, pretreatment of mice with either ketotifen (Figure 1A) or doxantrazole (Figure 1B)

prevented manipulation-induced gastroparesis and resulted in a significant decreased half-

emptying time (t1/2) back to laparotomy control values. In conjunction, ketotifen pretreatment

prevented the increase in gastric retention (Figure 1A and B). Although doxantrazole

pretreatment led to a normalized t1/2 , its effect was less effective compared with ketotifen

because gastric retention at 60 minutes was still significantly increased (Figure 1B).

Page 149: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

148

-L

treatmentsurgery

MPO

act

ivity

(U/m

g tis

sue)

ketoIM

0

20

40

60

ketoL

-L

doxIM

doxL

-IM

*

-IM

*

Figure 2. The increase in ileal myeloperoxidase (MPO) activity after surgery with intestinal manipula-tion is prevented by ketotifen or doxantrazole pretreatment. MPO activity was determined in whole homogenates of ileum, isolated 24 hours after surgery. The MPO activity after IM in mice pretreated with ketotifen vehicle is increased, but no increase is seen after pretreatment with mast cell stabilizer ketotifen (solid bars). Similarly, doxantrazole pretreatment prevented postoperative increase in MPO activity, compared with its respective vehicle treated control (grey bars). Treatment with ketotifen or doxantrazole had no effect on basal MPO activity in control animals. Asterisks indicate significant dif-ferences in ketotifen or doxantrazole treatment group using a 1-way ANOVA (P < 0.05), followed by Dunnett’s multiple comparison test. Data represent means ± SEM of 5–8 mice.

We next investigated whether the effect of mast cell stabilizers on normalizing postoperative

gastric emptying could be ascribed to an attenuation of the intestinal inflammatory response

to bowel surgery. Therefore, the inflammatory infiltrate in the intestine was quantified by

measuring MPO activity in intestinal homogenates 24 hours after intestinal manipulation.

Figure 2 shows that intestinal manipulation significantly increased intestinal MPO activity

in mice treated with vehicle. This increase in postoperative intestinal MPO activity was

prevented by pretreatment with ketotifen or doxantrazole, compared with their respective

vehicle-treated controls, indicating that mast cell degranulation is an important step in the

establishment of the leukocyte infiltrate. We subsequently analyzed the effect of mast cell

Page 150: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

7M

ast c

ell D

egra

nula

tion

Initi

ates

in P

osto

pera

tive

Ileus

149

stabilization on the inflammation of the intestinal muscularis by staining for MPO containing

leukocytes in muscularic whole-mount preparations. Figure 3 shows that, at 24 hours after

surgery, extensive leukocyte infiltration into the intestinal muscularis was detected in mice

that were treated with vehicle, corroborating earlier results9,11; however, pretreatment with

either ketotifen or doxantrazole significantly reduced the number of leukocytes infiltrating

the intestinal muscularis tissue.

In Vivo Mast Cell Degranulation in the Ileum Results in Leukocyte Infiltration and the Development of POITo evaluate further the importance of mast cell degranulation in initiating muscularic

inflammation and the development of POI, an isolated bowel segment was exposed to

the mast cell secretagogue C48/80.27 C48/80 has been shown to activate and degranulate

CTMC effectively.28 We studied whether the mast cell degranulation and the subsequent

muscularic inflammation elicited gastroparesis, similar to that seen after intestinal

manipulation. To this end, we measured gastric emptying 24 hours after selective exposure

of the ileum to C48/80 (Figure 4). C48/80 incubation led to a significant delay in gastric

emptying, compared with a similar treatment with vehicle (0.9% NaCl). Half-emptying

times, as well as gastric retention at 60 minutes after consumption of the test meal, were

significantly increased. Ketotifen pretreatment prevented the delay in gastric emptying

after C48/80 treatment Figure 4), implying that the gastroparesis developed as a result

of degranulation of CTMC. C48/80-induced mast cell degranulation resulted in a marked

leukocyte infiltration into the intestinal muscularis 24 hours after C48/80 exposure (Figure

5). The degree of muscular inflammation observed after this treatment was equal to that

24 hours after intestinal manipulation. In conjunction, the MPO activity in intestinal loops

exposed to C48/80 (Figure 6) was significantly increased, compared with loops exposed to

vehicle (0.9% NaCl).

Pretreatment of mice with ketotifen ablated the increase in MPO activity after C48/80

exposure but did not affect the MPO activity measured after treatment with saline (Figure

6), demonstrating that the increase in MPO activity seen after C48/80 treatment selectively

resulted from CTMC degranulation.

Page 151: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

150

Figure 3. The appearance of leukocyte infiltrates in ileal muscularis after intestinal manipulation is prevented by ketotifen or doxantrazole pretreatment. (A–D) Whole mount preparations of ileal intesti-nal muscularis tissue 24 hours after L (A), IM with ketotifen vehicle (B), IM with ketotifen pretreatment (C), and IM with doxantrazole pretreatment (D) are stained for MPO positive leukocytes. IM with either ketotifen or doxantrazole (not shown) vehicle pretreatment induced a massive influx of MPO-positive leukocytes to the ileal muscularis, compared with L (A and B). Pretreatment with ketotifen (C) or doxantrazole (D) prevented this influx of inflammatory cells. Preparations shown are representa-tive for 5–8 mice per treatment group. Bar is0.6 mm. (E) Shows that the significant increase in the number of MPO positive leukocytes per mm2 of muscularis tissue after IM with (ketotifen) vehicle pretreatment was prevented by ketotifen or doxantrazole pretreatment. Asterisk indicates significant difference using a 1-way ANOVA (P < 0.05), followed by Dunnett’s multiple comparison test. Values shown are the mean cell counts ± SEM of muscularis prepared from 5–8 mice.

Page 152: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

7M

ast c

ell D

egra

nula

tion

Initi

ates

in P

osto

pera

tive

Ileus

151

20

60

100re

lativ

e ga

stric

con

tent

(%)

20 60 100time (min)

saline

C48/80C48/80 + ketotifen

t (min)

32.6 ± 2.259.1 ± 10.4 *37.6 ± 7.9

salineC48/80C48/80

--+

ketotifenpretreated

intestinalexposure

30.4 ± 4.452.3 ± 7.4 *34.5 ± 5.3

Ret60min (%)

Figure 4. Intestinal exposure to C48/80 delays gastric emptying. A Gastric emptying curves, deter-mined by scintigraphic imaging of the abdomen after oral administration of solid caloric meal at 24 hours after exposure to C48/80 (solid circles), exposure to C48/80 after pretreatment with ketotifen (open circles), and L alone (squares). Exposure to C48/80 results in a delay in gastric emptying, which can be prevented by ketotifen pretreatment. Values are given as percentage of gastric content compared with the total abdominal region. Corresponding half-emptying time (t1/2) as well as the retention after 60 minutes. Ret60min is significantly increased after C48/80 exposure, compared with vehicle (saline) (B). Pretreatment with ketotifen restores both t1/2 as well as Ret60min back to normal. Values are means ± SEM of 8–12 mice per treatment group. Asterisks indicate significant differences at P < 0.05.

Page 153: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

152

Figure 5. (see fullcolor chapter 11) Mast cell degranulation results in infiltration of leukocytes in ileal muscularis. (A and B) Whole mount preparations of ileal intestinal muscularis tissue 24 hours after exposure to vehicle (0.9% NaCl) (A) or C48/80 (B) are stained for MPO-positive leukocytes. Exten-sive inflammatory infiltrates were observed after exposure to C48/80 but not saline. Preparations shown are representative for 6–8 mice per treatment group. Bar is 0.6 mm. Panel C shows that the number of MPO-positive leukocytes was significantly increased after incubation with C48/80, com-pared with incubation with vehicle. Asterisk indicates significant difference (P < 0.05). Values shown are the means ± SEM of 6–8 mice.

Page 154: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

7M

ast c

ell D

egra

nula

tion

Initi

ates

in P

osto

pera

tive

Ileus

153

10

30

50

ketotifen pretreatment08/84C08/84Cerusopxe lanitsetni -

-NaCl

-- +NaCl

+

MPO

act

ivity

(U/m

g tis

sue)

*

Figure 6. Mast cell degranulation elicits an increase in ileal MPO activity that can be prevented by ketotifen pretreatment. MPO activity was determined in whole homogenates of ileum isolated 24 hours after exposure to C48/80, or vehicle only (saline). The MPO activity after exposure to C48/80 is significantly increased, whereas exposure to saline did not affect MPO activity, irrespective of keto-tifen pretreatment. Note that the increase in MPO activity elicited by C48/80 exposure was prevented by ketotifen pretreatment. Asterisk indicates significant difference using a 1-way ANOVA (P < 0.05), followed by Dunnett’s multiple comparison test. Data represent means ± SEM of 6–8 mice.

Page 155: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

154

Mast Cell-Deficient Mice Are Resistant to Manipulation-Induced Muscularic InflammationTo confirm further that mast cells participate in the generation of the intestinal inflammation

that mediates POI, we performed abdominal surgery on Kit/Kitv mutant mice. These mice

have been shown to lack mast cells in all anatomical sites investigated.23 Indeed, no mast

cells could be identified in tissue sections of intestine, mesentery, or cytospins of peritoneal

fluid after staining with Toluidine blue (Figure 7, left panels). Intestinal manipulation

performed on Kit/WT congenic wild-type mice elicited mast cell degranulation indicated by

the increased level of MMCP-1 in their peritoneal lavage fluid measured 20 minutes after

surgery, compared with control laparotomy (13.0 ± 2.0 vs. 0.3 ± 0.2 ng/mL, respectively).

As expected, levels of peritoneal MMCP-1 levels were hardly detectable in Kit/Kitv mutant

mice and did not increase upon intestinal manipulation (0.2 ± 0.1 and 0.3 ± 0.2 ng/mL after

laparotomy and intestinal manipulation, respectively).

Page 156: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

7M

ast c

ell D

egra

nula

tion

Initi

ates

in P

osto

pera

tive

Ileus

155

The absence of mast cells led to a significant reduction in the manipulation-induced

inflammation of the intestine. Intestinal manipulation performed on Kit/WT congenic wild-

type mice resulted in an increased MPO activity into the intestinal tissue, compared with

laparotomy alone (Figure 8, solid bars). In Kit/Kitv mutant mice, however, MPO activity

was not significantly increased after intestinal manipulation. In concert, the number of

leukocytes infiltrating the intestinal muscularis after abdominal surgery performed on Kit/

Kitv mutant mice was significantly reduced compared with the number seen in Kit/WT wild-

type muscularis (Figure 9A and B).

Figure 7. (see fullcolor chapter 11) Reconstituted mast cells in Kit/Kitv mutant mice have a normal phenotypic appearance. (A and C) Mast cells were absent in Kit/Kitv small intestinal muscularis and Peyer’s patch (LM, longitudinal muscle layer; CM, circular muscle layer; PP, Peyer’s patch), as well as in peritoneal fluid (E). Sections of small intestinal muscularis (B and D) and peritoneal fluid (F) of Kit/Kitv mice reconstituted with cultured bone marrow-derived Kit/WT wild-type mast cells. The num-ber of mast cells recovered in reconstituted mice is similar to that in wild-type mice, and they have a normal histology and granule content (arrows). Giemsa staining. Sections are representative of 5 mice examined in each group. Bar is 75 μm.

Page 157: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

156

10

30

50

LKit/Kit v

IMKit/Kit v

IMKit/WT

MPO

act

ivity

(U/m

g tis

sue)

*

IMKit/Kit v

PBS

*

IMKit/Kit v

Kit/WT MC

Figure 8. Intestinal inflammation after intestinal manipulation depends on the presence of mast cells. MPO activity was determined in whole homogenates of ileum isolated 24 hours after L or IM. A sig-nificant increase in MPO activity and inflammation was observed after IM in wild-type mice but not in mast cell deficient Kit/Kitv mutants (solid bars). Reconstitution of Kit/Kitv mutant mice with cultured Kit/WT mast cells restored the granulocyte infiltration after intestinal manipulation to wild-type levels. Asterisks indicate significant differences (P < 0.05). Data represent means ± SEM of 5 mice.

Page 158: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

7M

ast c

ell D

egra

nula

tion

Initi

ates

in P

osto

pera

tive

Ileus

157

Figure 9. (see fullcolor chapter 11) Granulocyte infiltration into the intestinal muscularis after intestinal manipulation in mast cell deficient- and mast cell–reconstituted mice. (A–D) Whole mount preparations of ileal intestinal muscularis tissue 24 hours after IM stained for MPO-positive leukocytes. Extensive inflammatory infiltrates were observed after IM in Kit/WT mice (A), but the number was drastically reduced in Kit/Kitv mutant mice (B). Reconstitution of Kit/Kitv mutant mice with Kit/WT mast cells restored the inflammatory response to IM (D), whereas reconstitution with PBS did not (C). Preparations shown are the representative for 5 mice per treatment group. Bar is 0.6 mm. The number of MPO-positive leukocytes was significantly decreased after IM in Kit/Kitv mutant mice, compared with Kit/WT wild-type, whereas mast cell reconstitution restored the inflammatory response to IM. Asterisk indicates significant difference (P < 0.05). Values shown are the means ± SEM of 5 mice.

Page 159: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

158

Reconstitution of Mast Cells Restores Manipulation-Induced Intestinal Inflammation in Mast Cell-Deficient MiceTo demonstrate directly the role of mast cells in the manipulation-induced inflammatory

response in the intestinal muscularis, mast cell populations were restored in Kit/Kitv mice

by adoptive transfer of cultured mast cells derived from congenic Kit/WT wild-type mice. If

mast cell deficiency alone would account for the lack of muscularic inflammation observed

in Kit/Kitv mice, reconstitution of the mast cell population in these animals should restore

the inflammation to the level of Kit/WT wild-type mice. To this end, we performed mast cell

reconstitution in Kit/Kitv recipients by IP injection of cultured bone marrow-derived mast

cells to repair the mast cell deficit. Reconstitution of Kit/Kitv mice gave rise to phenotypically

(Figure 7, right panels) and quantitatively normal mast cell populations in peritoneal lavage

fluid, mesentery, and intestine 10–12 weeks after transplantation, confirming earlier

reports.23 We performed abdominal surgery on mast cell reconstituted Kit/Kitv mice and

investigated the intestinal MPO activity and leukocyte infiltration 24 hours after surgery. In

Figure 8, it is shown that MPO activity was significantly increased after bowel manipulation

in reconstituted animals, compared with non-reconstituted, age-matched, mast cell

deficient mice (Figure 8, gray bars). Analysis of the MPO containing leukocytes in the

intestinal muscularis stained by muscularic whole-mount staining (Figure 9, panels C and

D) showed that intestinal manipulation gave rise to a significant increase in the number of

infiltrating leukocytes in mast cell-reconstituted mice, whereas surgery performed on non-

reconstituted Kit/Kitv mice injected with PBS did not.

Page 160: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

7M

ast c

ell D

egra

nula

tion

Initi

ates

in P

osto

pera

tive

Ileus

159

DiscussionIn previous studies, it has been established that inflammation of the small intestinal muscularis

resulting from bowel manipulation is the main contributor to the prolonged phase of POI.9,11

The mechanism leading to the inflammatory response to bowel manipulation however, is

not known. Intense activation of visceral afferents, i.e., because of mechanical stretch, can

result in the local release of sensory neurotransmitters, especially substance P and CGRP.

These neuromediators have pro-inflammatory effects in that their release has been shown

to elicit a neurogenic inflammation at the activated tissue site.29,30 In our current model,

visceral afferents most likely are triggered to release these neuropeptides during intestinal

manipulation. Mast cells have been shown to be in close contact with visceral afferent

nerve terminals,31 and secreted neuropeptides can directly activate mast cells.32–34 We

therefore investigated the role of mast cell activation in the recruitment of the manipulation-

induced inflammation. First, we showed that bowel manipulation indeed increased the level

of peritoneal mast cell protease mMCP-1. Second, stabilization of mast cells using either

doxantrazole or ketotifen prevented this increase and prevented the intestinal inflammation

and delayed gastric emptying following bowel manipulation as well. The involvement of

mast cells in this process was further demonstrated by exposure of a segment of small

intestine to the mast cell-degranulating compound C48/80. Similar to bowel manipulation,

local mast cell degranulation induced by C48/80 resulted in inflammation of the exposed

bowel segment and delayed gastric emptying. These data clearly illustrate a crucial role of

mast cells in the generation of the inflammation following intestinal manipulation.

To confirm further the requirement of mast cells in the induction of the manipulation-induced

intestinal inflammation, we conducted experiments in mast cell-deficient Kit/Kitv animals.

These mice displayed a significantly reduced inflammation after bowel manipulation. The

mast cell deficiency of these mice is due to mutations in the c-kit receptor gene, which

impairs the development of functional mast cells derived from the bone marrow. Because

of the fact that this mutation also affects other cell lineages, such as red blood cells and

melanocytes,35 these mice are mildly anemic, although immune responses have been

described to be generally similar to wild-type mice.36 In addition, the lack of a functional

c-kit receptor affects proper development of the network of the interstitial cells of Cajal,37

Page 161: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

160

resulting in a disturbed gastrointestinal motility in these mice. Loss of these cells has been

associated with aberrations in gastric emptying.38 Therefore, data of gastric emptying were

not obtained from these mice.

To rule out the possibility that resistance of mast cell-deficient mice to manipulation-induced

inflammation is due to anemia or other defects resulting from the W mutation in Kit/Kitv

mice apart from mast cell deficiency, adoptive transfer of immature mast cells derived from

bone marrow cells of congenic normal Kit/WT mice was performed to repair selectively

the mast cell deficiency of the Kit/Kitv recipients. The final maturation and phenotype of

bone marrow-derived cultured mast cells transferred to mast cell-deficient Kit/Kitv mice

has been shown to be determined by the tissue in which mast cells are located.39 Hence,

mast cell-reconstituted Kit/Kitv mice differ only from Kit/Kitv mice in their presence of mast

cells. We found the mast cell populations in reconstituted mice histologically normal, and

normal numbers of mast cells were recovered from intestine, stomach, mesentery, and

peritoneum, which confirms earlier reports.23 We observed that mast cell reconstitution

of Kit/Kitv mice with mast cells derived from wild-type animals restored the manipulation-

induced inflammatory response in the intestinal muscularis back to wild-type levels,

showing that neutrophil infiltration and subsequent muscularic inflammation triggered by

intestinal manipulation are mast cell dependent.

MMC in the intestinal mucosa or CTMC in mesentery, serosa, and lamina propria have distinct

phenotypes and functions.14 Treatment with doxantrazole, which stabilizes both MMC and

CTMC,20,40 and ketotifen, a stabilizer of mainly CTMC,20 were both effective in preventing

the occurrence of muscular inflammation that follows bowel surgery. Unexpectedly, our

data also indicate that doxantrazole tended to be less effective in reducing postoperative

gastroparesis, although both stabilizers were equally potent in attenuating mMCP-1

release in the peritoneal cavity. Nevertheless, we must conclude that CTMC are involved

early in the process of the recruitment of inflammatory cells following bowel manipulation.

In particular, CTMC, and not MMC, respond to C48/80.41 C48/80 exposure mimicked the

manipulation-induced inflammation and gastroparesis, again suggesting that only CTMC

are involved in the initiation of the manipulation-induced inflammation. Paradoxically

however, intestinal manipulation elicited an increase in the level of mMCP-1, a soluble

chymase derived from MMC and not CTMC.14 These 2 observations indicate that both

Page 162: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

7M

ast c

ell D

egra

nula

tion

Initi

ates

in P

osto

pera

tive

Ileus

161

MMC and CTMC degranulate as a result of intestinal manipulation. Our observation that

CTMC degranulation using C48/80 results in an inflammation in the muscularis externa, i.e.,

a site anatomically distinct from the mesentery, implies that CTMC (for instance adhering

to the intestinal serosa) can easily exert proinflammatory effects on surrounding intestinal

tissue.

Kalff et al. have previously suggested that the intestinal inflammation observed after

manipulation results from activation of resident macrophages, in rodents11 as well as in

humans,42 possibly through the enhanced expression of LFA-1.11 The mechanism by which

these macrophages may be activated, however, has not been studied. One likely possibility

is that macrophages are activated through proinflammatory mast cell mediators released,

such as TNF-α.43 In addition, other mast cell-derived mediators, such as histamine,44

prostaglandines, and tryptase,45 can orchestrate inflammation, possibly via activation of

macrophages. To what extent mast cells activate resident macrophages or vice versa

cannot be concluded from our data but is currently the subject of ongoing studies.

In conclusion, our findings demonstrate that mast cells play an essential role in the genesis

of the muscularic inflammation mediating POI. Furthermore, we showed that mast cell

stabilization resulted in shortening the period of POI following bowel surgery. Mast cell

stabilizing agents are commonly used in the treatment of asthma and allergic disorders,46

and mast cell stabilizers have been shown in animal models22,47 and humans48,49 to attenuate

the severity of active colitis. Based on our data, clinical studies evaluating the effect of mast

cell stabilization as a possible treatment for POI are certainly warranted.

Page 163: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

162

Reference ListPrasad M, Matthews JB. Deflating postoperative ileus. Gastroenterology 1999;117:489–492.1. Livingston EH, Passaro EP Jr. Postoperative ileus. Dig Dis Sci 1990;35:121–132.2. De Winter BY, Boeckxstaens GE, De Man JG, Moreels TG, Herman AG, Pelckmans PA. Effect 3. of adrenergic and nitrergic blockade on experimental ileus in rats. Br J Pharmacol 1997;120: 464–468.Barquist E, Bonaz B, Martinez V, Rivier J, Zinner MJ, Tache Y. Neuronal pathways involved in 4. abdominal surgery-induced gastric ileus in rats. Am J Physiol 1996;270:R888–R894.Plourde V, Wong HC, Walsh JH, Raybould HE, Tache Y. CGRP antagonists and capsaicin on 5. celiac ganglia partly prevent postoperative gastric ileus. Peptides 1993;14:1225–1229.Raybould HE, Kolve E, Tache Y. Central nervous system action of calcitonin gene-related pep-6. tide to inhibit gastric emptying in the conscious rat. Peptides 1988;9:735–737.Zittel TT, Reddy SN, Plourde V, Raybould HE. Role of spinal afferents and calcitonin gene-re-7. lated peptide in the postoperative gastric ileus in anesthetized rats. Ann Surg 1994;219:79–87.Bonaz B, Tache Y. Corticotropin-releasing factor and systemic capsaicin-sensitive afferents 8. are involved in abdominal surgeryinduced Fos expression in the paraventricular nucleus of the hypothalamus. Brain Res 1997;748:12–20. de Jonge WJ, van den Wijngaard RM, The FO, ter Beek ML, Bennink RJ, Tytgat GN, Buijs 9. RM, Reitsma PH, van Deventer SJ, Boeckxstaens GE. Postoperative ileus is maintained by intestinal immune infiltrates that activate inhibitory neural pathways in mice. Gastroenterology 2003;125:1137–1147.Kalff JC, Buchholz BM, Eskandari MK, Hierholzer C, Schraut WH, Simmons RL, Bauer AJ. Bi-10. phasic response to gut manipulation and temporal correlation of cellular infiltrates and muscle dysfunction in rat. Surgery 1999;126:498–509. Kalff JC, Carlos TM, Schraut WH, Billiar TR, Simmons RL, Bauer AJ. Surgically induced leuko-11. cytic infiltrates within the rat intestinal muscularis mediate postoperative ileus. Gastroenterol-ogy 1999;117:378–387.Kalff JC, Cicalese L, Exner B, Schraut WH, Bauer AJ. Role of phagocytes in causing dysmotil-12. ity after each stage of small bowel transplantation. Transplant Proc 1998;30:2568.Moriwaki K, Fujii K, Yuge O. Protein exudation induced by manipulation of the intestines and 13. mesentery during laparotomy in rat. A study of the mechanism of “third space” loss. In Vivo 1997;11:325–327.Miller HR, Pemberton AD. Tissue-specific expression of mast cell granule serine proteinases 14. and their role in inflammation in the lung and gut. Immunology 2002;105:375–390Biedermann T, Kneilling M, Mailhammer R, Maier K, Sander CA, Kollias G, Kunkel SL, Hultner 15. L, Rocken M. Mast cells control neutrophil recruitment during T-cell-mediated delayed-type hypersensitivity reactions through tumor necrosis factor and macrophage inflammatory protein 2. J Exp Med 2000;192:1441–1152. Galli SJ, Gordon JR, Wershil BK. Cytokine production by mast cells and basophils. Curr Opin 16. Immunol 1991;3:865–872. Chen R, Ning G, Zhao ML, Fleming MG, Diaz LA, Werb Z, Liu Z. Mast cells play a key role in 17. neutrophil recruitment in experimental bullous pemphigoid. J Clin Invest 2001;108:1151–1158.Kanwar S, Kubes P. Mast cells contribute to ischemia-reperfusion-induced granulocyte infiltra-18. tion and intestinal dysfunction. Am J Physiol 1994;267:G316–G321.Williams CM, Galli SJ. Mast cells can amplify airway reactivity and features of chronic inflam-19. mation in an asthma model in mice. J Exp Med 2000;192:455–462.Pearce FL, Befus AD, Gauldie J, Bienenstock J. Mucosal mast cells. II. Effects of anti-allergic 20. compounds on histamine secretion by isolated intestinal mast cells. J Immunol 1982;128: 2481–2486.Brown JF, Chafee KA, Tepperman BL. Role of mast cells, neutrophils and nitric oxide in 21. endotoxin-induced damage to the neonatal rat colon. Br J Pharmacol 1998;123:31–38.

Page 164: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

7M

ast c

ell D

egra

nula

tion

Initi

ates

in P

osto

pera

tive

Ileus

163

Eliakim R, Karmeli F, Okon E, Rachmilewitz D. Ketotifen ameliorates capsaicin-augmented 22. acetic acid-induced colitis. Dig Dis Sci 1995;40:503–509.Kitamura Y, Go S, Hatanaka K. Decrease of mast cells in W/W23. v mice and their increase by bone marrow transplantation. Blood 1978;52:447–452.Wershil BK, Mekori YA, Murakami T, Galli SJ. 125I-fibrin deposition in IgE-dependent imme-24. diate hypersensitivity reactions in mouse skin. Demonstration of the role of mast cells using genetically mast cell-deficient mice locally reconstituted with cultured mast cells. J Immunol 1987;139:2605–2614.Bennink RJ, De Jonge WJ, Symonds EL, Van Den Wijngaard RM, Spijkerboer AL, Benninga 25. MA, Boeckxstaens GE. Validation of gastric-emptying scintigraphy of solids and liquids in mice using dedicated animal pinhole scintigraphy. J Nucl Med 2003;44: 1099–1104.Symonds EL, Butler RN, Omari TI. Assessment of gastric emptying in the mouse using the 26. [13C]-octanoic acid breath test. Clin Exp Pharmacol Physiol 2000;27:671–675.Koibuchi Y, Ichikawa A, Nakagawa M, Tomita K. Binding of active components of compound 27. 48/80 to rat peritoneal mast cells. Eur J Pharmacol 1985;115:171–177.Jaffery G, Coleman JW, Huntley J, Bell EB. Mast cell recovery following chronic treatment with 28. compound 48/80. Int Arch Allergy Immunol 1994;105:274–280.Quinlan KL, Song IS, Naik SM, Letran EL, Olerud JE, Bunnett NW, Armstrong CA, Caughman 29. SW, Ansel JC. VCAM-1 expression on human dermal microvascular endothelial cells is directly and specifically up-regulated by substance P. J Immunol 1999;162: 1656–1661.Smith CH, Barker JN, Morris RW, MacDonald DM, Lee TH. Neuropeptides induce rapid ex-30. pression of endothelial cell adhesion molecules and elicit granulocytic infiltration in human skin. J Immunol 1993;151:3274–3282.Williams RM, Berthoud HR, Stead RH. Vagal afferent nerve fibres contact mast cells in rat 31. small intestinal mucosa. Neuroimmunomodulation 1997;4:266–270.Ansel JC, Brown JR, Payan DG, Brown MA. Substance P selectively activates TNF-alpha gene 32. expression in murine mast cells. J Immunol 1993;150:4478–4485.Suzuki R, Furuno T, McKay DM, Wolvers D, Teshima R, Nakanishi M, Bienenstock J. Direct 33. neurite-mast cell communication in vitro occurs via the neuropeptide substance P. J Immunol 1999;163: 2410–2415.Theoharides TC, Singh LK, Boucher W, Pang X, Letourneau R, Webster E, Chrousos G. 34. Corticotropin-releasing hormone induces skin mast cell degranulation and increased vas-cular permeability, a possible explanation for its proinflammatory effects. Endocrinology 1998;139:403–413.Wershil BK, Tsai M, Geissler EN, Zsebo KM, Galli SJ. The rat c-kit ligand, stem cell factor, 35. induces c-kit receptor-dependent mouse mast cell activation in vivo. Evidence that signaling through the c-kit receptor can induce expression of cellular function. J Exp Med 1992;175:245–255.Wershil BK, Furuta GT, Wang ZS, Galli SJ. Mast cell-dependent neutrophil and mononuclear 36. cell recruitment in immunoglobulin E-induced gastric reactions in mice. Gastroenterology 1996;110:1482–1490.Huizinga JD, Thuneberg L, Kluppel M, Malysz J, Mikkelsen HB, Bernstein A. W/kit gene re-37. quired for interstitial cells of Cajal and for intestinal pacemaker activity. Nature 1995;373:347–349.Ordog T, Takayama I, Cheung WK, Ward SM, Sanders KM. Remodeling of networks of intersti-38. tial cells of Cajal in a murine model of diabetic gastroparesis. Diabetes 2000;49:1731–1739.Kitamura Y, Morii E, Ogihara H, Jippo T, Ito A. Mutant mice: a useful tool for studying the devel-39. opment of mast cells. Int Arch Allergy Immunol 2001;124:16–19.Francois A, Ksas B, Aigueperse J, Griffiths NM. The recovery of the neurally evoked secre-40. tory response of rat colonic mucosa after irradiation is independent of mast cells. Radiat Res 2002;157:266–274. Befus AD, Pearce FL, Gauldie J, Horsewood P, Bienenstock J. Mucosal mast cells. I. Isolation 41. and functional characteristics of rat intestinal mast cells. J Immunol 1982;128:2475–2480.

Page 165: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

164

Kalff JC, Turler A, Schwarz NT, Schraut WH, Lee KK, Tweardy DJ, Billiar TR, Simmons RL, 42. Bauer AJ. Intra-abdominal activation of a local inflammatory response within the human mus-cularis externa during laparotomy. Ann Surg 2003;237:301–315.Bissonnette EY, Enciso JA, Befus AD. Inhibitory effects of sulfasalazine and its metabolites on 43. histamine release and TNF-α production by mast cells. J Immunol 1996;156:218–223.Torres R, de Castellarnau C, Ferrer LL, Puigdemont A, Santamaria LF, de Mora F. Mast cells 44. induce upregulation of P-selectin and intercellular adhesion molecule 1 on carotid endothelial cells in a new in vitro model of mast cell to endothelial cell communication. Immunol Cell Biol 2002;80:170–177.Vergnolle N. Proteinase-activated receptor-2-activating peptides induce leukocyte rolling, adhe-45. sion, and extravasation in vivo. J Immunol 1999;163:5064–5069.Slater JW, Zechnich AD, Haxby DG. Second-generation antihistamines: a comparative review. 46. Drugs 1999;57:31–47.Eliakim R, Karmeli F, Okon E, Rachmilewitz D. Ketotifen effectively prevents mucosal damage 47. in experimental colitis. Gut 1992;33:1498–1503.Marshall JK, Irvine EJ. Ketotifen treatment of active colitis in patients with 5-aminosalicylate 48. intolerance. Can J Gastroenterol 1998;12:273–275.Jones NL, Roifman CM, Griffiths AM, Sherman P. Ketotifen therapy for acute ulcerative colitis 49. in children: a pilot study. Dig Dis Sci 1998;43:609–615.

Page 166: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

7M

ast c

ell D

egra

nula

tion

Initi

ates

in P

osto

pera

tive

Ileus

165

Page 167: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

8

Page 168: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

8Cha

pter

8Intestinal Handling Induced

Mast Cell Activation and

Inflammation in Human

Postoperative Ileus

Frans O. The, Roelof J. Bennink, Willem M Ankum,

Marrije R. Buist, Olivier R.C. Busch,

Dirk J. Gouma, Sicco van der Heide,

René M. van den Wijngaard, Wouter J. de Jonge,

Guy E. Boeckxstaens

Gut 2008; 57: 33-40

Page 169: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

168

AbstractBackground & Aims: Murine postoperative ileus results from intestinal inflammation trig-

gered by manipulation-induced mast cell activation. As its extent depends on the degree

of handling and subsequent inflammation, we hypothesise that the faster recovery after

minimal invasive surgery results from decreased mast cell activation and impaired intes-

tinal inflammation. Objective: to quantify mast cell activation and inflammation in patients

undergoing conventional and minimal invasive surgery. Methods: 1)Mast cell activation

(i.e. tryptase release) and pro-inflammatory mediator release were determined in perito-

neal lavage fluid obtained on consecutive time-points during open, laparoscopic and trans-

vaginal gynaecological surgery. 2)LFA-1, ICAM-1 and iNOS mRNA as well as leukocyte

influx were quantified in non-handled and handled jejunal muscle specimens collected dur-

ing biliary reconstructive surgery. 3)Intestinal leukocyte influx was assessed by 99mTc la-

belled leukocyte SPECT-CT scanning before and after abdominal or vaginal hysterectomy.

Results: 1)Intestinal handling during abdominal hysterectomy resulted in an immediate

release of tryptase followed by enhanced IL-6 and IL-8 levels. None of the mediators rose

during minimal invasive surgery except for a slight increase in IL-8 during laparoscopic

surgery. 2)Jejunal mRNA transcription for ICAM-1 and iNOS as well as leukocyte recruit-

ment were increased after intestinal handling. 3)Leukocyte scanning 24hrs after surgery

revealed increased intestinal activity after abdominal but not after vaginal hysterectomy.

Conclusions: This study demonstrates that intestinal handling triggers mast cells activa-

tion and inflammation associated with prolonged postoperative ileus. These results may

partly explain the faster recovery after minimal invasive surgery and encourage future clini-

cal trials targeting mast cells to shorten postoperative ileus.

Page 170: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

8M

ast C

ell i

nduc

ed In

flam

mat

ion

in H

uman

Pos

tope

rativ

e Ile

us

169

PBackgroundPostoperative ileus, characterized by a lack of coordinated motility of the entire

gastrointestinal tract leads to increased morbidity and prolonged hospitalisation1,2 and

represents a substantial socio-economical burden. In the US alone, the additional annual

healthcare expenses related to this condition have been estimated to surpass 1 billion

dollars3. At present, treatment is rather disappointing and limited to predominantly supportive

measures4.

The introduction of minimal invasive surgical techniques (e.g. laparoscopy) has fastened

postoperative recovery significantly5. This major improvement is believed to result from

minimal wound trauma and decreased release of stress hormones6,7. In addition, it is

becoming increasingly clear that intestinal inflammation is a key event in the pathogenesis

of postoperative ileus. In rats, the degree of gut paralysis is directly proportional to the

degree of intestinal handling and inflammation8. This inflammation leads to local impaired

muscle contractility9 and the activation of an adrenergic inhibitory neural pathway10.

Reduction of inflammatory cell influx accomplished by blockade of adhesion molecules

shortens postoperative ileus10,11 and further underscores the importance of this handling

induced inflammation. As minimal invasive surgery implies limited handling of the intestine,

faster recovery of motility may result from an impaired influx of inflammatory cells.

Although the exact mechanism remains unclear, we previously showed that mast cells

play a pivotal role in triggering the inflammatory process. In mice, intestinal handling led

to degranulation of mast cells with increased levels of mouse mast cell protease-1 in

peritoneal lavage fluid. In contrast, W/Wv mice, deficient of mast cells, failed to develop an

intestinal muscle inflammation in response to manipulation of a bowel loop. Reconstitution

of W/Wv mice with mast cells from wild type animals restored the handling induced

inflammatory response, clearly demonstrating the importance of mast cells. Activation of

resident macrophages has also been demonstrated, possibly secondary to influx of luminal

bacteria during a brief episode of increased mucosal permeability12. To what extent mast

cell activation is the trigger leading to increased mucosal permeability and macrophage

activation remains to be determined.

Page 171: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

170

At present the evidence supporting the importance of inflammation in human is rather

scarce13, and data on the relationship between the degree of inflammation and clinical

outcome are lacking. In addition, although we provided convincing evidence for a crucial

role of mast cell degranulation in mice, no data are available in man. In the present study,

therefore, we studied whether intestinal manipulation leads to mast cell degranulation

and inflammation in patients undergoing conventional or minimal invasive surgery and

hypothesised that clinical recovery is determined by the degree of manipulation induced

mast cell activation and inflammation.

Page 172: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

8M

ast C

ell i

nduc

ed In

flam

mat

ion

in H

uman

Pos

tope

rativ

e Ile

us

171

Patients and MethodsParticipantsBetween December 2003 and July 2005 a total of 44 patients were enrolled in 3 clinical

research protocols. The physical condition and co-morbidity of potential participants

was assessed during pre-assessment at the outpatient clinic of the department of

anaesthesiology, which is part of the standard pre-operative work-up. The American Society

of Anesthesiologists-Physical Status classification (ASA-PS)14,15 was used and comprises

a scale from 1 to 6 in which 1 equals a normal healthy patient; 2 equals a patient with

mild systemic disease; 3 equals a patient with severe systemic disease; 4 is a patient

with severe systemic disease that is a constant threat to life; 5 equals a moribund patient

who is not expected to survive without the operation and 6 equals a declared brain-dead

patient whose organs are being removed for donor purposes14. In the present study, only

patients categorized as ASA-PS 1 to 3 were asked to participate. In addition, patients were

screened for the following exclusion criteria: intra-abdominal inflammation, pre-operative

radiation therapy and the use of anti-inflammatory or mast cell stabilizing drugs. Patients

were included after informed consent was obtained.

Study designThe activation of mast cells and the inflammatory mediator response to intestinal handling

were evaluated in protocol 1. Pro-inflammatory gene transcription and leukocyte recruitment

within the handled intestinal muscle layer were studied in protocol 2. The occurrence of

manipulation induced leukocyte recruitment in relation to clinical recovery of bowel function

and duration of hospital admission was evaluated in study protocol 3. All protocols were

evaluated and approved by the Medical Ethical Review Board of the Academic Medical

Center, Amsterdam, the Netherlands.

AnaesthesiaTo correct for the influence of used anesthetic technique and medication, patients were

subjected to standardized peri-operative care according to our anaesthesiologist’s protocol.

In short, patients were pre-medicated with paracetamol 1000mg and lorazepam 1mg on

the evening before surgery and approximately 2hrs before surgery. Induction of general

Page 173: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

172

anaesthesia was attained with propovol 2-2.5mg/kg; fentanyl 1.5-3μg/kg; rocuronium

0.6mg/kg. Anesthesia was maintained using et. 0.8% isoflurane. Postoperative pain

medication was introduced after last sampling in protocol 1 and 2 and administered similar

for all patients according to our anesthesiologist’s pain protocol.

Protocol 1: Mast cell activation and inflammatory mediator release during abdominal surgeryPeritoneal lavage fluid samples were collected from 18 patients, either undergoing an

abdominal hysterectomy (n=6), a laparoscopic resection of an adnexum (n=6) or a trans-

vaginal hysterectomy (n=6). Three consecutive lavages were performed in each individual

patient. The first lavage sample was collected immediately after opening of the peritoneum

(basal). A second lavage sample was collected immediately after abdominal inspection

and first gentle small intestinal handling (early). The final lavage sample was collected at

the end of the procedure (late). As the intestine is not handled in the trans-vaginal group,

only two lavages were performed in the trans-vaginal hysterectomy group (i.e. basal and

late sampling). The systemic release of mediators in response to abdominal surgery was

assessed in 2 blood samples (abdominal hysterectomy only), the first sample taken before

induction of general anaesthesia (1day prior to surgery) and the second sample taken

at the end of the surgical procedure, i.e. just before closure of the abdominal cavity. The

harvested fluid and serum were used to measure the release of tryptase, Tumor Necrosis

Factor (TNF)-α Interleukin (IL)-1β, IL-6 and IL-8 in relation to surgical handling. The

abdominal lavage was performed using 100ml of warm (42°C) sterile 0.9% NaCl solution,

which was sprinkled gently onto the small intestine and its mesentery. After approximately

30 seconds, peritoneal fluid (between 20 and 40 ml) was collected using a 22 French Foley

catheter (Bard Limited, West Sussex, England) connected to a 50ml catheter tip syringe.

Protocol 2: Regulatory gene transcription and leukocyte influx upon intestinal handlingJejunal muscle specimens were used to quantify regulatory gene transcription and

assess the degree of inflammation. Full thickness biopsies were obtained from patients

undergoing biliary reconstructive surgery. This specific procedure was chosen because of

its considerable length, providing at least sufficient time for gene transcription to occur16.

Two consecutive jejunal tissue samples were collected from 10 patients. The first specimen

Page 174: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

8M

ast C

ell i

nduc

ed In

flam

mat

ion

in H

uman

Pos

tope

rativ

e Ile

us

173

was collected at the beginning of the procedure and had not been touched by the surgeon

until resection. The second tissue specimen, exposed to the usual handling during surgery,

was collected approximately 3 hrs thereafter. Following mucosa removal, both specimens

were partitioned (5mm2 segments) and snap frozen in liquid nitrogen in the operating

theatre and stored at -80°C.

Protocol 3: Abdominal leukocyte recruitment and clinical recoveryAbdominal leukocyte Single Photon Emission Computed Tomography (SPECT) CT scans

were performed in 16 gynaecological patients to quantify the leukocyte recruitment in

response to surgical handling. Eight patients undergoing an abdominal hysterectomy were

compared with 8 patients undergoing a vaginal hysterectomy. In each patient a reference

(basal) leukocyte scintigraphy was performed on the day of admission, 24 hrs prior to

surgery. A second leukocyte scintigraphy was performed on the first postoperative day, i.e.

approximately 24 hrs after surgery. Clinical recovery was assessed until hospital discharge

(see methods for detailed description).

Page 175: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

174

MethodsTryptase releaseTryptase concentrations were assessed at the routine clinical laboratory of the Department

of Allergy, University Medical Center, Groningen, the Netherlands. Total tryptase

(α-protryptase and β-tryptase) concentration was measured in peripheral blood and lavage

fluid samples using a commercial fluoro-immunoenzyme assay (FIA) (Pharmacia Uppsala,

Sweden)17.

Cytokine and chemokine releaseCytokine levels were determined by cytometric bead array (BD PharMingen, San Diego,

CA, USA). In short, 5μl of each test sample was mixed with 5μl of mixed capture beads

and 5μl of human phycoerythrin (PE) detection reagents consisting of PE-conjugated anti-

human IL-1β, TNF-α, IL-6 and IL8. These mixtures were incubated at room temperature in

dark for 3 hrs, washed and resuspended in 300ul wash buffer. Acquisition was performed

on a FACSCalibur using a high throughput-sampling interface (BD Biosciences, Sunnyvale,

CA, USA). Generated data were analyzed using CBA software (BD PharMingen, San

Diego, CA, U SA) and interpolated from corresponding standard curves generated using

the mixed cytokine standard provided by the supplier18.19.

Real Time Reverse Transcription-Polymerase Chain ReactionTissue specimens were homogenized and total RNA was extracted using Trizol

(Invitrogen, Carlsbad, CA, USA). The total RNA fractions were treated with DNAse

and reverse-transcribed using Superscript II (Invitrogen, Carlsbad, CA, USA).

cDNA (150ng) was subjected to 45 cycles of lightcycler PCR (FastStartDNA

Masterplus SYBR Green; Roche, Basel Switzerland). The following primers

were used: LFA-1 antisense 5’-GACCCAAGTGCTCTCAGGAA-3’ and sense 5’-

AGGAGCACTCCACTTCATGC-3’; ICAM-1 antisense 5’-CATAGAGACCCCGTTGCCTA-3’

and sense 5’-GGGTAAGGTTCTTGCCCACT- 3’; iNOS antisense 5’-TGGAAGCGGTA

ACAAAGGAGA- 3’ and sense 5’ CGATGCACAGCTGAGTGAAT- 3’; GAPDH antisense 5’-

CGACCACTTTGTCAAGCTCA-3’ and sense 5’-AGGGGAGATTCAGTGTGGTG-3’. PCR

quantification was performed by a linear regression method using the Log(fluorescence)

Page 176: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

8M

ast C

ell i

nduc

ed In

flam

mat

ion

in H

uman

Pos

tope

rativ

e Ile

us

175

per cycle number20 and normalized for GAPDH housekeeping gene expression. In each

individual patient the late sample value was expressed as fold increase of the early control

sample value.

ImmunocytochemistryImmunocytochemical staining was performed on peritoneal cell cytospins obtained from

the harvested abdominal lavage fluid. In short, spins containing 1x105 cells were fixed in

carnoy’s fixation fluid (60% ethanol, 30% chloroform and 10% glacial acidic acid) for 30

min at room temperature and washed with TBST (0.1%). Non-specific binding of antibody

was blocked by incubation with TBS containing 10% normal goat serum for 20 min. Spins

were incubated with anti-tryptase antibodies (mouse anti-human, 1:250) (Chemicon,

Temecula, CA, USA) for 2 hrs at room temperature. Goat-anti mouse alexa-488 was used

as secondary antibody (Molecular probes, Invitrogen, Carlsbad, CA, USA). After final

washing, the spins were mounted using Vectashield mounting medium containing 5μg/ml

DAPI (Vector Laboratories, Burlingame, CA, USA).

Semi-quantitative evaluation of the degree of intestinal muscle inflammationHandled and non-handled jejunal muscle sections were used to assess the extent of

inflammation. Leukocyte infiltration was visualized by myeloperoxidase (MPO) staining as

described previously(11). After 10min fixation in ice-cold acetone, transverse frozen section

(8 μ) were incubated for 10 minutes with 3-amino-9-ethyl carbazole (Sigma, St. Louis,

MO) as a substrate, dissolved in sodium acetate buffer (pH 5.0) to which 0.01% H2O2 was

added10. To evaluate the degree of inflammation, unmarked myeloperoxidase stained early

and late collected section from 10 patients were scored independently by 3 observers

(TK, OW and RVDW). A semi-quantitative scoring scale from 0 to 4 was utilized; 0 being

non-inflamed, 1=very mildly inflamed, 2=mildly inflamed, 3= inflamed and 4 being clearly

inflamed. The mean of 3 scores, calculated for each segment, was used for statistical

analysis (Wilcoxon signed rank test).

In-vivo quantification of leukocyte recruitmentWhite blood cells (WBC) were labeled using technetium-99m hexamethylpropyleneamine

oxime (99mTc-HMPAO) (Ceretec, GE Health, Eindhoven, The Netherlands) according to

Page 177: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

176

the consensus protocol for leukocyte labelling21. The harvested WBC fraction of 100ml of

blood labeled with an average of 450 ± 10 MBq of 99mTc-HMPAO was reinjected into the

patient. Sixty min later, a SPECT scan of the abdomen was performed (GE Millennium

Hawkeye, GE Healthcare, Den Bosch, the Netherlands) followed by a low dose CT-scan

without contrast on the same gantry. CT data were used for attenuation correction and as

an anatomical reference for region of interest (ROI) analysis. After data acquisition, images

were processed on an Entegra workstation (GE Healthcare, Den Bosch, The Netherlands)

using attenuation corrected iterative reconstruction and analyzed on a Hermes workstation

(Nuclear Diagnostics, Stockholm, Sweden). Five consecutive abdominal SPECT slices

were summed and ROI’s were drawn around small intestine and lumbar spine at the level

of the ileac crest. Small bowel uptake of leukocytes was calculated as an uptake ratio

expressed as a fraction of bone marrow activity, similar to analysis of leukocyte uptake

assessment in inflammatory bowel disease22. The small bowel uptake ratio determined

prior to surgery was considered as basal leukocyte activity. The relative percentage of

difference in leukocyte activity 24h after surgery was calculated using the following formula:

(postoperative small bowel ratio/preoperative small bowel ratio)*100%.

Clinical evaluationAll patients received standard postoperative medical care according to the ward accustomed

care protocol. Patients were visited by the research physician once daily until discharge

to assess postoperative clinical recovery of bowel function (time of first flatus and time of

first defecation). Patients were discharged when the following criteria were met: normal

urinary-tract function, spontaneous defecation, tolerance of oral fluid and solid food intake,

adequate pain relief with oral analgesics and adequate mobilization and self-support.

Statistical analysisStatistical analysis was performed using SPSS 12.02 software for Windows. Data were

non-parametrically distributed and expressed as median values and inter quartile range

or median increase compared to basal values. In protocol 1, all serum but only vaginal

hysterectomy lavage samples were analysed using a Wilcoxon signed rank test for 2 paired

samples. For all other lavage sample-series (consisting of 3 samples) a Friedman’s two

way analysis of variance was applied. When a statistical difference was observed, a Mann-

Whitney test used to identify the specific sample(s) of significant difference. In protocol

Page 178: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

8M

ast C

ell i

nduc

ed In

flam

mat

ion

in H

uman

Pos

tope

rativ

e Ile

us

177

2, the quantitative PCR data and the semi-quantitative inflammation data were analyzed

using a Wilcoxon signed rank test. In protocol 3, leukocyte recruitment was analyzed using

the Wilcoxon signed rank test. Clinical data were analyzed with a Mann-Whitney test for

independent samples. P-values <0.05 was considered statistically significant.

Page 179: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

178

ResultsPatient demographicsEighteen patients participated in study protocol 1, 6 in each surgical intervention group.

Overall mean age was 47 years, range 21 to 70 (trans–vaginal: 52 years, range 43 to

70; laparoscopy: 36 years, range 21 to 49; laparotomy: 49 years, range 44 to 53). The

indications for surgery in this patient population were leyomyomata (n=8), prolaps (n=4) or

a benign ovarian tumour (n=6). In study protocol 2, jejunal tissue samples were collected

from 10 patients (6 male, mean age 42 years, range 32 to 53) who underwent biliary

reconstructive surgery because of iatrogenic biliary tract injury. Study protocol 3 involved

16 patients; 8 patients underwent an abdominal hysterectomy (mean age 50 years, range

42 to 70) and 8 patients underwent a vaginal hysterectomy (mean age 55 years, range 42

to 66). The indications for surgery were uterine leiomyomata in the abdominal hysterectomy

patient group and uterine prolapse (n=4), leyomyomata (n=3) and primary dysmenorrhoea

(n=1) in the vaginal hysterectomy group.

Study protocol 1:Mast cell activation and inflammatory response during abdominal surgery

To assess the activation of mast cells in response to intestinal handling, the expression and

release of tryptase, a pre-stored mast cell specific protease23, was analyzed. Peritoneal

lavage fluid harvested during abdominal surgery (laparotomy) contained a distinct mast

cell population, as illustrated by the number of tryptase positive cells in fig. 1a. In the

basal lavage sample collected immediately after opening of the peritoneal cavity, the

basal median tryptase concentration was 5.2 (InterQuartile Range (IQR) 2.7-11.3) μg/l.

Tryptase release was significantly increased to a median concentration of 23.1 (IQR 15.1-

46.9) μg/l, (p=0.02) in early samples taken after gentle palpation of the small intestines,

necessary to allow inspection of the pelvic organs. In the late sample taken at the end

of surgery, tryptase levels had increased even further (late: median concentration 51.7

(IQR 25.8-90.2) μg/l, n=6, p=0.002) (fig.1b). In contrast, neither laparoscopic nor trans-

vaginal intra-peritoneal surgery (n=6 in both types of surgery) elicited a significant mast cell

response (fig. 1c-d). To evaluate possible release of mast cell mediators in the systemic

circulation, we also determined pre- and postoperative serum tryptase concentrations in

Page 180: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

8M

ast C

ell i

nduc

ed In

flam

mat

ion

in H

uman

Pos

tope

rativ

e Ile

us

179

Figure 1 (see fullcolor chapter 11)a) Peritoneal cells collected in late lavage flu-id stained for tryptase (in green). Cell nuclei were counterstained with DAPI (bleu). Indi-vidual patient tryptase concentrations during b) open surgery, c) laparoscopic surgery and d) trans-vaginal surgery; measured in lavage fluid collected immediately after opening of the peritoneal cavity (basal), after first palpa-tion of the small intestine during inspection of pelvic organs (early) and at the end of the procedure (late). A Wilcoxon signed rank test (vaginal samples) and Friedman’s two way analysis of variance (laparoscopic and laparotomy samples) were used to deter-mine statistical significance. Tryptase levels increased significant in patient undergoing a laparotomy (n=6, p=0.002) in contrast to the laparoscopic (n=6, p=0.5) or vaginal (n=6, p=0.06) approach. Note that no “early” la-vage was performed in patients undergoing trans-vaginal surgery. Dotted line represents median change in tryptase concentration of all 6 patients

D

ar scop

late du ing surge

trans-vaginal

basal early late0

25

50

75

100

125

150

time during surgery

tryp

tase

con

c. (

g/L)C laparoscopy

basal early late0

25

50

75

100

125

150

time during surgery

tryp

tase

con

c. (

g/L)

2 0

B

D

laparotomy

basal early late0

25

50

75

100

125

150

†*

*

time during surgery

tryp

tase

con

c. (

g/L)

apa oscopy

du ng su g

trans-vagina

bas ear a

2

5

7

10

12

15

time dur g surger

y

. (g/

C laparoscop

bas ear la

2

5

7

10

12

time duri g surger

y

(/

Page 181: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

180

the laparotomy group (n=6 patients). Serum tryptase levels did not increase and remained

within the normal range of 1 to 11.4 μg/l24 (pre-operative median concentration 4.1 (IQR

2.8-7.5) μg/l and postoperative 1.6 (IQR 1.3-3.5) μg/l respectively). The release of the

pro-inflammatory cytokines TNF-α IL-1β, IL-6 and the chemokine IL-8 was analyzed in

the same peritoneal lavage samples. Gentle handling of the intestine during laparotomy

did not lead to an immediate increase in any of these mediators. However, at the end of

the surgical procedure, IL-6 and IL-8 were increased significantly (table 1). In laparoscopic

treated patients, intra-peritoneal IL-8, but not IL-6, was increased, but not as profound as

in the laparotomy group (table 1). On the other hand, transvaginal surgery did not affect

any of the measured cytokines and chemokines. TNF-α and IL-1β levels did not change

upon first handling or at the end of any of the types of surgery evaluated. The serum levels

of the studied inflammatory mediators remained unaltered (median increase compared to

pre-operative for: TNF-α: 0.0 (IQR 0.0-0.0) pg/ml; IL-1β: 0.0 (IQR 0.0-35.3) pg/ml; IL-6: 4.2

(IQR 0.0-11.7) pg/ml; IL-8: 0.9 (IQR 0.0-310.4) pg/ml).

Table 1Pro-inflammatory mediator release in lavage fluids collected immediately after opening of the perito-neal cavity (basal), after first palpation of the small intestine during inspection of pelvic organs (early) and at the end of the surgical procedure (late). A Wilcoxon signed rank test (vaginal samples) and Friedman’s two way analysis of variance (laparoscopic and laparotomy samples) were used to deter-mine statistical significance. TNF-α and IL-1β did not change during surgery. IL-8 increased signifi-cant at the end of laparoscopic as well as open (laparotomy) surgery. IL-6 only increased at the end of a laparotomy. None of the pro-inflammatory proteins increased in the trans-vaginal surgery group. Note that no “early” lavage was performed in patients undergoing trans-vaginal surgery.

Table 1: Inflammatory mediator release during surgery

treatment group laparotomy (n=6) laparoscopy (n=6) trans-vaginal (n=6)

TNF- 0.0 (0.0-3.4) 0.0 (-6.6-0.0) 0.0 (0.0-0.0)

IL-1 0.0 (-1.8-18.1) -2.5 (-3.3-0.0) 0.2 (0.0-0.8)

IL-6 135.6 (4.2-5130.0) 1 6.1 (1.3-15.2) 1.5 (-0.4-4.0)

IL-8 114.2 (32.9-208.7) 2 28.9 (1.3-166.5) 1 0.8 (-2.8-6.0)

median increase of mediator concentration in late vs. basal lavage sample collected during indicated

type of surgery (pg/ml) with (IQR) 1 p=0.02; 2 p=0.006

able 2: Qua tita ive ge e tra sc iption nalys s.

0

Page 182: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

8M

ast C

ell i

nduc

ed In

flam

mat

ion

in H

uman

Pos

tope

rativ

e Ile

us

181

Study protocol 2Regulatory gene transcription upon intestinal handling

Recruitment of leukocytes to the muscularis propria strongly depends on the upregulation

of adhesion molecules and the synthesis of pro-inflammatory proteins. Therefore, ICAM-1,

LFA-1 and iNOS gene expression was determined in muscle specimens collected during

abdominal surgery. As the synthesis of functional proteins requires several hours16, mRNA

quantification was used to evaluate the kinetics of these inflammatory proteins in jejunal

muscle tissue. As shown in Table 2, iNOS and ICAM-1 levels were significantly increased

after intestinal handling (table 2) In contrast LFA-1 remained unchanged.

Degree of intestinal muscle inflammation upon intestinal handling.

Histological evaluation of leukocyte recruitment was performed before and after

surgical handling on the same tissue specimens used for gene-transcription analysis.

Myeloperoxidase was stained to visualize leukocyte recruitment in response to intestinal

handling in transverse sections of the jejunal muscularis propria. Non-handled early samples

contained only a small number of leukocytes in the muscle layer (fig 2 upper left panel). In

contrast, routinely handled late specimens showed a marked extravasation of inflammatory

cells (fig. 2 upper right panel), confirmed by semi-quantitative evaluation (fig.2 lower panel).

These recruited leukocytes predominantly reside in and around the vasculature of the

handled intestinal muscle layers as is illustrated in figure 2b. This extravasation marks the

ongoing inflammatory process.

e 1: Inflamm tory mediat r r a durin s gery

.6 .2 0. . . - 0

1 4 2 32.9 208 7 8 9 1.3-166.5 0.8 (-2.8 6.0)

an increase of m d ator concen rati n n late vs. s va e sample col ec ed du ing in c ted

f surg (pg ml) wi h (IQR) 1 p=0 2; p=0 00

Table 2: Quantitative gene transcription analysis.

Gene median fold increase of gene expression

LFA-1 0.9 (0.3-20.0)

ICAM-1 3.3 (1.3-139.9) 1

iNOS 3.3 (0.7-20.0) 2

median fold increase of gene transcription in late (handled) vs. early (non-handled)

jejunal muscle layer with (IQR) (n= 10 patients)

1 p=0.017; 2 p=0.022

Table 2Relative increase of mRNA expression in manipulated small intestinal muscle tissue compared to non-handled control specimens. A Wilcoxon signed rank test was used to determine statistical dif-ferences. The relative increase was significant for iNOS (median fold increase 3.3 (IQR 0.7-20.0), p=0.022) and ICAM1 (median fold increase 3.3 (IQR 1.3-139.9), p=0.017).

Page 183: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

182

early late

0

1

2

3

4

p=0.005

degr

ee o

f inf

lam

mat

ion

(sca

le 0

-4)

Figure 2Handling induced leukocyte infiltration of the jejunal muscularis propria, visualized by myeloperoxi-dase staining in early non-handled (upper panel, left) and late handled tissue segments (upper panel right). Note the ongoing extravasation, illustrated by the predominant peri-vascular localization of leukocytes in the handled late tissue sample. Semi-quantitative evaluation of handling induced leuko-cyte recruitment is depicted in the lower panel (scale 0= non-inflamed through 1=very mildly inflamed, 2=mildly inflamed, 3=inflamed to 4= clearly inflamed). Early non-handled (median score: 1 (IQR 1-2)) vs. Late handled (median score: 3 (IQR 2-4)), n= paired samples from10 patients, p=0.005 tested with a Wilcoxon signed rank test. The dotted line represents the median increase in intestinal muscle inflammation of all 10 patients.

early

100x

dee

late

100x

Page 184: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

8M

ast C

ell i

nduc

ed In

flam

mat

ion

in H

uman

Pos

tope

rativ

e Ile

us

183

Study protocol 3Abdominal leukocyte recruitment 24hrs after open and minimal invasive hysterectomy

In vivo leukocyte recruitment in response to intestinal handling was investigated by 99mTc

labeled leukocyte imaging. Abdominal leukocyte influx was assessed on 5 consecutive

leukocyte-SPECT images at the level of the ileac crest and compared with that of the bone

marrow(22). The change in leukocyte activity before compared to after surgery showed

no increase in the vaginal hysterectomy group (median % of activity before compared

to after surgery 91% (IQR 84-102), n=8). In the abdominal hysterectomy group however

leukocyte recruitment was significantly increased to a median of 127% of the pre-operative

abdominal activity ((IQR 113-148), n=8, p=0.01) (fig 3). To determine the exact anatomical

location, plain CT-images were made immediately after SPECT imaging. The region in

which the enhanced leukocyte activity was observed coincided with small intestinal loops

and its mesentery, as shown in figure 4.

75

100

125

150

175

200 *

abdominalhysterectomy

vaginalhysterectomy

post

-ope

rativ

e le

ukoc

yte

activ

ity (%

of b

asal

)

Figure 3Quantification of postoperative leukocyte recruitment to the small intestinal region expressed as per-centage (%) of the preoperative scan. A significant increase (Wilcoxon signed rank test) in intestinal leukocyte activity was observed after an abdominal hysterectomy (median % of preoperative scan 127% (IQR 113-148), n=8, p=0.01), but not after a vaginal hysterectomy (median % of preoperative scan 91% (IQR 84-102), ns, n=8).

Page 185: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

184

Figure 4 (see fullcolor chapter 11)Representative example of leukocyte SPECT-CT imaging 24 hrs before (left column) and after (right column) an abdominal hysterectomy was performed. a) Coronary SPECT overview slide with ana-tomical references: (1) liver, (2) bladder, (3) ileac spine and (4) lumbar vertebral-range between which quantification was performed. b) transverse CT-slide and c) corresponding SPECT image at same position in quantification range, visualizing increased leukocyte activity in the abdominal region (ar-rows). Finally, d) transfers CT- and SPECT overlay showing the specific leukocyte activity in the small intestine (arrow heads).

Page 186: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

8M

ast C

ell i

nduc

ed In

flam

mat

ion

in H

uman

Pos

tope

rativ

e Ile

us

185

Clinical recovery after open and minimal invasive hysterectomy

In conjunction with the assessed leukocyte recruitment, clinical recovery was also evaluated.

Time until first flatus did not differ significantly between the two patient groups. First bowel

movement and duration of hospital admission however were significantly prolonged after

abdominal hysterectomies compared to the vaginal procedure (table 3).

Table 3Patient demographics and clinical recovery data from patients undergoing a vaginal hysterectomy or an abdominal hysterectomy assessed in protocol 3. To identify potential confounders in clinical pa-rameters and to test significant difference a Mann-Whitney test was performed. Age and ASA-score did not differ between the two patient populations. Time till first defecation (1 p=0.02) and time until discharge 2p=0.001) were both significantly prolonged in patients undergoing an abdominal hyster-ectomy when compared to those undergoing a vaginal hysterectomy. All data are depicted as mean and (range).

Table 3: Clinical evaluation of post-operative recovery

treatment group abdominal

hysterectomy

vaginal

hysterectomy

mean age (years) 50 (range 42-70) 55 (range 42-66)

mean ASA-PS* 2 (range 1-3) 1 (range 1-2)

mean time of surgery (min) 182 (range 130-298) 150 (range 113-179)

mean time until first flatulence (days) 2 (range 2-3) 1 (range 1-2)

mean time until first defecation (days) 4 (range 4-5) 1 2 (range 2-3)

mean time until discharge (days) 8 (range 7-8) 2 4 (range 4-5)

*ASA-PS: American Society of Anaesthesiologists-Physical Status (scale 1 (being a normal

healthy patients) to 6 (being a patient declared brain-dead) see methods section for detailed

description) 1 p=0.02, 2 p=0.001

Page 187: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

186

DiscussionInflammation of the muscularis propria following surgical manipulation of the intestine is

increasingly recognized to postpone the recovery of gastrointestinal motility. Animal studies

indeed have revealed that prevention of this inflammatory process, either by antibodies

or antisense oligonucleotides to the adhesion molecule ICAM-1, macrophage inactivation

or COX-2 inhibition enhances gastrointestinal transit and shortens postoperative ileus11,

25-27. Recently, we demonstrated that mast cell activation plays an important role in this

process and may be one of the first steps triggering the inflammatory response. Intestinal

manipulation indeed induces the immediate activation of mast cells leading to increased

levels of the murine mast cell proteinase-1 in the abdominal cavity25. Three hours later,

inflammatory mediators such as MIP-2, MIP-1α TNFα and IL-6 can be detected27, 28 which

on their turn enhance the expression of adhesion molecules such as ICAM-111, recruitment

of leukocytes and inflammation of the intestine.

In the present study, we investigated whether this cascade of events also plays a role in the

pathogenesis of human postoperative ileus. We found that gentle palpation of the intestines

during first inspection of the pelvic organs resulted in the instantaneous intra-peritoneal

release of tryptase, a mast cell specific protease23, in patient undergoing a laparotomy.

This increase in tryptase increased even further towards the end of the procedure and

was accompanied by an increase in IL-6 and IL-8. As the latter is known to be released in

response to mast cell activation and initiates leukocyte recruitment via ICAM-129, we also

determined ICAM-1 and iNOS mRNA in intestinal tissue that was handled at the start of a

surgical procedure, but was only removed approximately three hours later. In addition to

an upregulation of ICAM-1 and iNOS, the number of inflammatory cells was significantly

increased in these late tissue samples compared to untouched specimen harvested at the

beginning of the procedure. Interestingly, leukocytes were localized predominantly around

blood vessels in both the serosa and the muscularis propria, partly adhering to the endothelial

lining marking the ongoing recruitment and extravasation in this early stage of inflammation.

Kalff et al. also reported an intestinal inflammatory response during abdominal surgery in

patients13. To assess the degree of inflammation in a later stage, we also performed 99mTc

labelled leukocyte SPECT scanning 24 hrs after surgery. Using this technique, we showed

Page 188: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

8M

ast C

ell i

nduc

ed In

flam

mat

ion

in H

uman

Pos

tope

rativ

e Ile

us

187

increased intra-abdominal activity compared to the pre-operative baseline scan in patients

subjected to an abdominal hysterectomy. As the actual resection, performed in the pelvic

region, did not comprise any gastrointestinal organs, this observed increase in leukocyte

activity can not be explained by the primary surgical trauma. Clearly, leukocytes could

reside anywhere in the abdominal cavity and may not be restricted to the intestinal wall.

The additional CT scanning however showed that the increased leukocyte activity observed

with the SPECT scans coincided with intestinal loops. When the uterus was resected

transvaginally, a surgical approach that leaves the intestines largely untouched, no such

increase was observed, indirectly suggesting that the intestinal inflammation is triggered by

intestinal manipulation. From these data we conclude that also in man, manipulation of the

intestine during surgery leads to mast cell degranulation and a local inflammatory process,

which, like in our animal model, plays an important role in postoperative hypomotility.

In rodents, the extent of gastrointestinal hypomotility or ileus is proportionally related to the

degree of intestinal handling and subsequent inflammation8. As intestinal manipulation is

minimal in laparoscopic surgery, one might argue that the degree of mast cell degranulation

and the subsequent inflammatory response in the intestine will be less and thus may

contribute to the faster clinical recovery observed after minimal invasive surgery. To test

this hypothesis, tryptase and inflammatory mediators were quantified during 2 minimal

invasive surgical procedures, i.e. laparoscopic and trans-vaginal hysterectomy. In contrast

to gentle handling during open surgery, no mast cell degranulation or increase of IL-6

was observed in the peritoneal lavage fluid. Only IL-8 levels were increased, although

less profound compared to laparotomy. Moreover, during trans-vaginal surgery, leaving the

intestines largely untouched, none of the evaluated parameters increased. These findings

underscore that the degree of intestinal handling to a large extent determines the degree

of mast cell activation and the subsequent inflammatory response. The latter was further

confirmed by the 99mTc labelled leukocyte SPECT scanning 24 hrs after surgery showing

increased intra-abdominal activity in patients subjected to an abdominal hysterectomy but

not in patients who underwent a trans-vaginal hysterectomy. Finally, clinical recovery in our

study was significantly delayed after abdominal compared to vaginal hysterectomy, a finding

in line with previous clinical studies showing faster postoperative gastrointestinal recovery

after minimal invasive surgery6, 30-35. It should be emphasized though that differences in

postoperative pain medication, especially opioids, may have contributed to the delay in

Page 189: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

188

normalisation of gastrointestinal motility36,37. In the current study, however, postoperative

analgesia in both patient groups was provided according to a standardized postoperative

pain protocol, making this explanation less likely. Therefore, the observation that delayed

clinical recovery is associated with increased influx of radio-labeled leukocytes indirectly

adds to the hypothesis that the degree of intestinal handling, mast cell degranulation and

subsequent inflammation determine the duration of postoperative ileus.

A drawback of this study is that the study-protocols were conducted in different groups of

patients. Ideally, the same patient cohort should have been studied to better understand

the causative association between mast cell degranulation and the subsequently observed

inflammatory responses upon intestinal handling. Especially as mRNA levels of ICAM-1

and iNOS peak only 2 to 24hrs after stimulation16, 38 a long-lasting surgical procedure had to

be chosen in order to allow the detection of the upregulation of these inflammatory markers

in response to intestinal handling. Therefore patients undergoing biliary reconstructive

surgery were selected instead.

Our current findings may have important clinical implications. First, they clearly illustrate

that manipulation of the intestine should be limited whenever possible in order to reduce

the release of mast cell mediators and limit postoperative intestinal inflammation. This

knowledge should urge further development of minimal invasive surgical or even endoscopic

techniques to minimise intestinal handling. Second, if mast cell degranulation is indeed

an important initial step in the pathophysiology of postoperative ileus in man, mast cells

may represent an important therapeutic target. As we previously showed reduction of

postoperative ileus by mast cell stabilisation in our mouse model, our current findings in

humans warrant further studies evaluating the effect of a mast cell stabilising agent in

patients.

Page 190: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

8M

ast C

ell i

nduc

ed In

flam

mat

ion

in H

uman

Pos

tope

rativ

e Ile

us

189

Reference ListCollins TC, Daley J, Henderson WH et al. Risk factors for prolonged length of stay after major 1. elective surgery. Ann Surg 1999;230(2):251-9.Longo WE, Virgo KS, Johnson FE et al. Risk factors for morbidity and mortality after colectomy 2. for colon cancer. Dis Colon Rectum 2000;43(1):83-91.Prasad M, Matthews JB. Deflating postoperative ileus. Gastroenterology1999;117(2):489-92.3. Kehlet H, Holte K. Review of postoperative ileus. Am J Surg 2001;182(5A Suppl):3S-10S.4. Schwenk W, Haase O, Neudecker J et al. Short term benefits for laparoscopic colorectal resec-5. tion. Cochrane Database Syst Rev 2005;(3):CD003145.Chen HH, Wexner SD, Iroatulam AJ et al. Laparoscopic colectomy compares favor-6. ably with colectomy by laparotomy for reduction of postoperative ileus. Dis Colon Rectum 2000;43(1):61-5.Glaser F, Sannwald GA, Buhr HJ et al. General stress response to conventional and laparo-7. scopic cholecystectomy. Ann Surg 1995;221(4):372-80.Kalff JC, Schraut WH, Simmons RL et al. Surgical manipulation of the gut elicits an intestinal 8. muscularis inflammatory response resulting in postsurgical ileus. Ann Surg 1998;228(5):652-63.Kalff JC, Carlos TM, Schraut WH et al. Surgically induced leukocytic infiltrates within the rat 9. intestinal muscularis mediate postoperative ileus. Gastroenterology 1999;117(2):378-87.de Jonge WJ, van den Wijngaard RM, The FO et al. Postoperative ileus is maintained by 10. intestinal immune infiltrates that activate inhibitory neural pathways in mice. Gastroenterology 2003;125(4):1137-47.The FO, de Jonge WJ, Bennink RJ et al. The ICAM-1 antisense oligonucleotide ISIS- 3082 11. prevents the development of postoperative ileus in mice. Br J Pharmacol 2005.Schwarz NT, Beer-Stolz D, Simmons RL et al. Pathogenesis of paralytic ileus: intestinal ma-12. nipulation opens a transient pathway between the intestinal lumen and the leukocytic infiltrate of the jejunal muscularis. Ann Surg 2002;235(1):31-40.Kalff JC, Turler A, Schwarz NT et al. Intra-abdominal activation of a local inflammatory re-13. sponse within the human muscularis externa during laparotomy. Ann Surg 2003;237(3):301-15.Saklad M. Grading of patients for surgical procedures. Anesthesiology 1941;2:281-4.14. New Classification of Physical Status. American Society of Aesthesiologists,Inc. Anesthesiol-15. ogy 1963;24:111.Yan HC, Juhasz I, Pilewski J et al. Human/severe combined immunodeficient mouse chimeras. 16. An experimental in vivo model system to study the regulation of human endothelial cell-leuko-cyte adhesion molecules. J Clin Invest 1993;91(3):986-96.Schwartz LB, Kepley C. Development of markers for human basophils and mast cells. J Allergy 17. Clin Immunol 1994;94(6 Pt 2):1231-40. Tarnok A, Hambsch J, Chen R et al. Cytometric bead array to measure six cytokines in twenty-18. five microliters of serum. Clin Chem 2003;49(6 Pt 1):1000-2. Chen R, Lowe L, Wilson JD et al. Simultaneous Quantification of Six Human Cytokines 19. in a Single Sample Using Microparticle-based Flow Cytometric Technology. Clin Chem 1999;45(9):1693-4.Ramakers C, Ruijter JM, Deprez RH et al. Assumption-free analysis of quantitative realtime 20. polymerase chain reaction (PCR) data. Neurosci Lett 2003;339(1):62-6.Roca M, Martin-Comin J, Becker W et al. A consensus protocol for white blood cells labelling 21. with technetium-99m hexamethylpropylene amine oxime. International Society of Radiolabeled Blood Elements (ISORBE). Eur J Nucl Med 1998;25(7):797-9.Weldon MJ, Masoomi AM, Britten AJ et al. Quantification of inflammatory bowel disease activity 22. using technetium-99m HMPAO labelled leucocyte single photon emission computerised tomog-raphy (SPECT). Gut 1995;36(2):243-50.Hogan AD, Schwartz LB. Markers of mast cell degranulation. Methods 1997;13(1):43-52.23.

Page 191: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

190

Schwartz LB, Bradford TR, Rouse C et al. Development of a new, more sensitive immunoassay 24. for human tryptase: use in systemic anaphylaxis. J Clin Immunol 1994;14(3):190-204.de Jonge WJ, The FO, van der CD et al. Mast cell degranulation during abdominal surgery initi-25. ates postoperative ileus in mice. Gastroenterology 2004;127(2):535-45.Schwarz NT, Kalff JC, Turler A et al. Prostanoid production via COX-2 as a causative mecha-26. nism of rodent postoperative ileus. Gastroenterology 2001;121(6):1354-71.de Jonge WJ, van der Zanden EP, The FO et al. Stimulation of the vagus nerve attenu-27. ates macrophage activation by activating the Jak2-STAT3 signaling pathway. Nat Immunol 2005;6(8):844-51.Wehner S, Behrendt FF, Lyutenski BN et al. Inhibition of macrophage function prevents intesti-28. nal inflammation and postoperative ileus in rodents. Gut 2006.Compton SJ, Cairns JA, Holgate ST et al. The role of mast cell tryptase in regulating endothe-29. lial cell proliferation, cytokine release, and adhesion molecule expression: tryptase induces expression of mRNA for IL-1 beta and IL-8 and stimulates the selective release of IL-8 from human umbilical vein endothelial cells. J Immunol 1998;161(4):1939-46.Isik-Akbay EF, Harmanli OH, Panganamamula UR et al. Hysterectomy in obese women: a 30. comparison of abdominal and vaginal routes. Obstet Gynecol 2004;104(4):710-4. Veldkamp R, Kuhry E, Hop WC et al. Laparoscopic surgery versus open surgery for colon can-31. cer: short-term outcomes of a randomised trial. Lancet Oncol 2005;6(7):477-84.Graber JN, Schulte WJ, Condon RE et al. Relationship of duration of postoperative ileus to 32. extent and site of operative dissection. Surgery 1982;92(1):87-92.Huilgol RL, Wright CM, Solomon MJ. Laparoscopic versus open ileocolic resection for Crohn’s 33. disease. J Laparoendosc Adv Surg Tech A 2004;14(2):61-5.Bohm B, Milsom JW, Fazio VW. Postoperative intestinal motility following conventional and 34. laparoscopic intestinal surgery. Arch Surg 1995;130(4):415-9.Milsom JW, Hammerhofer KA, Bohm B et al. Prospective, randomized trial comparing laparo-35. scopic vs. conventional surgery for refractory ileocolic Crohn’s disease. Dis Colon Rectum 2001;44(1):1-8.Miedema BW, Johnson JO. Methods for decreasing postoperative gut dysmotility. Lancet Oncol 36. 2003;4(6):365-72.Bauer AJ, Boeckxstaens GE. Mechanisms of postoperative ileus. Neurogastroenterol Motil 37. 2004;16 Suppl 2:54-60.Yoo HS, Rutherford MS, Maheswaran SK et al. Induction of nitric oxide production by bo-38. vine alveolar macrophages in response to Pasteurella haemolytica A1. Microb Pathog 1996;20(6):361-75.

Page 192: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

8M

ast C

ell i

nduc

ed In

flam

mat

ion

in H

uman

Pos

tope

rativ

e Ile

us

191

Page 193: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

9

Page 194: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

9Cha

pter

9Mast Cell Stabilization as

Treatment of Postoperative

Ileus: a Pilot Study

Frans O. The, Marrije R. Buist,

Aaltje Lei, Roelof J. Bennink,

Jan Hofland, René M. van den Wijngaard,

Wouter J. de Jonge, Guy E. Boeckxstaens

submitted for publication

Page 195: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

194

AbstractBackground & Aim: Postoperative ileus is mediated by intestinal inflammation resulting

from manipulation-induced mast cell activation. Therefore, mast cell stabilization may

represent a new therapeutic approach to shorten postoperative ileus. Aim: To study the

effect of ketotifen, a mast cell stabilizer, on postoperative gastrointestinal transit in patients

who underwent abdominal surgery. Methods: In this pilot study, 60 patients undergoing

major abdominal surgery for gynecological malignancy with standardized anesthesia were

randomized to treatment with ketotifen (4 or 12mg) or placebo. Patients were treated for

6 days starting 3 days prior to surgery. Gastric emptying of liquids, selected as primary

outcome parameter, was measured 24 hrs after surgery using scintigraphy. Secondary

endpoints were, scintigraphically assessed, colonic transit represented as geometrical

center of activity (segment 1=cecum to 7=stool) and clinical parameters. Results: Gastric

retention 1 hr after liquid intake was significantly reduced by 12mg (median 3% (1-7),

p=0.01), but not by 4mg ketotifen (18 % (3-45), p=0.6) compared to placebo (16 %

(5-75)). Twenty-four hr colonic transit in placebo was 0.8 (0.0-1.1) vs. 1.2 (0.2-1.4) colon

segments in 12 mg ketotifen group (p=0.07). Abdominal cramps improved significantly in

patients treated with12mg ketotifen, whereas other clinical parameters were not affected.

Conclusion: Ketotifen significantly improves gastric emptying and showed a tendency

to improvement of colonic transit after abdominal surgery. These results warrant further

exploration of mast cell stabilizers as putative therapy for postoperative ileus.

Page 196: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

9M

ast C

ell S

tabi

lizat

ion

in Il

eus:

a R

ando

miz

ed T

rial

195

PBackgroundPostoperative ileus, characterized by generalized gastrointestinal hypomotility is a major

determinant of prolonged hospitalization after extensive abdominal surgery1. The annual

costs related to ileus have been estimated to exceed $1,000,000,000 in the US, illustrating

its socio-economical impact2. Until recently, neurogenic inhibition of gastrointestinal motility

was considered as main pathophysiological mechanism underlying postoperative ileus.

Animal experiments indeed revealed activation of adrenergic and non-adrenergic non-

cholinergic inhibitory pathways during and shortly after abdominal surgery3-5. To overcome

this inhibitory input, treatment so far has mainly focused on prokinetic drugs, such as

metoclopramide6-8, cisapride9-11 or erythromycin12, 13 with however disappointing results14, 15.

Therefore, there is a large need for other more efficient therapeutic strategies.

Recently, we and others have shown that local inflammation of the intestine triggered

by handling of bowel loops during surgery plays a crucial role in the pathogenesis of

postoperative ileus. In rodents, abdominal surgery indeed leads to influx of inflammatory

cells, approximately 4 to 6 hours after the intestinal segment has been manipulated. This

local inflammatory response not only leads to impaired neuromuscular function of the

affected intestinal segment, but also activates an adrenergic neural pathway inhibiting

the motility of the entire gastrointestinal tract. Most importantly, prevention of the influx

of inflammatory cells by for example blocking adhesion molecules such as ICAM-1

restores gastric emptying and intestinal transit16, 17, indicating the eminent role of this local

inflammatory response in the pathophysiology of postoperative ileus. Also in man, we and

others provided evidence that abdominal surgery triggers an inflammatory response in

intestinal tissue resected at the end of the procedure18, 19. Moreover, using SPECT imaging,

we were able to provide in vivo evidence for influx of radiolabeled leukocytes into the

intestine after open hysterectomy but not after laparoscopic hysterectomy18.

One of the initial steps attracting inflammatory cells to the site of manipulation is mast

cell degranulation. Intestinal handling triggers the release of mast cell mediators both in

rodents20 and man18, whereas W/Wv mice, deficient of mast cells, fail to develop intestinal

inflammation in response to bowel manipulation. Reconstitution of these animals with

Page 197: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

196

mast cells from their wild type littermates restores the occurrence of manipulation-induced

inflammation. Finally, pretreatment of mice with the mast cell stabilizing agents ketotifen

and doxantrazole prevents the occurrence of inflammation and normalizes postoperative

gastric emptying, suggesting that mast cell stabilization may be an attractive alternative

approach to treat postoperative ileus.

To investigate this hypothesis and to prove the concept that interference with the mast

cell – inflammation sequence indeed improves postoperative gastrointestinal motility, we

designed a double blind placebo controlled randomized pilot study evaluating the effect of

ketotifen on postoperative gastrointestinal transit.

Page 198: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

9M

ast C

ell S

tabi

lizat

ion

in Il

eus:

a R

ando

miz

ed T

rial

197

MethodsStudy subjectsThe present study is a randomized, double blind, placebo controlled, single center proof

of principle study conducted in the Academic Medical Center (AMC), Amsterdam, the

Netherlands. This study was approved by the Medical Ethical Committee of the AMC.

Patients (18 – 80 years of age), scheduled to undergo a radical hysterectomy, debulking

of ovarian malignancy, or an oncological explorative laparotomy were invited to participate.

The exclusion criteria were: 1) evident intra-abdominal inflammation (diagnosed by imaging

and/or laboratory test results), 2) use of anti-allergic drugs, 3) use of anti-inflammatory

pharmaca during the first 3 days after surgery, 4) use of laxatives and/or prokinetic agents

during the first 3 post-operative days, 5) colostomy or ileostomy, 6) intestinal resection as

part of the surgical procedure, 7) American Society of Anesthesiologists physical-health

status(ASA-PS)21 > III.

After written informed consent was obtained, patients were randomized to receive treatment

with placebo, ketotifen 4mg or ketotifen 12mg in 2 daily oral doses. In order to avoid side

effects such as sedation, the drug was gradually introduced (1/4 of the full dose on day 1,

1/2 of the full dose on day 2 and the full dose from day 3 until day 6). Randomization was

performed according to a 2:2:2 block ratio.

Study protocolTreatment was started 3 days prior to surgery and was continued until the second

postoperative day. Patients were admitted to the hospital 1 day prior to surgery. On the

evening before the operation the patients were pre-medicated with lorazepam 1mg orally,

followed by 1mg on the day of operation, to which then paracetamol 1000mg was added

(table 1). Anesthesia, analgesia, peri-operative intravenous (iv) fluids and respiratory

support were standardized according to a pre-defined protocol (table 1). The nasogastric

decompression tube was removed in the recovery room or on the ward the morning of the

first postoperative day. Postoperative analgesia was attained with paracetamol 500mg 6

times a day, orally. Non-steroidal anti-inflammatory drugs (NSAID’s) and/or tramadol were

Page 199: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

198

added on demand when iv. or epidural analgesia was ceased, however, NSAID’s were not

allowed before post-operative day 4.

On post-operative day 1 (24hrs after surgery), patients were asked to drink 100mL of

diethylenetriaminepentaacetate (111In-DTPA) labeled tap water. One, 24 and 48 hrs after

intake scintigraphical scans of the abdomen were performed (see section gastrointestinal

transit studies for details). Clinical recovery was monitored and symptoms were noted until

hospital discharge (see section data collection for details).

Page 200: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

9M

ast C

ell S

tabi

lizat

ion

in Il

eus:

a R

ando

miz

ed T

rial

199

mai

nten

ance

top-

up

test

dos

e

leve

l

VT6-

8ml/k

g e

nd-

title

CO 2

4-4.

5kPa

PEEP

5cm

H 2OFi

O240

% (a

ir-O 2)

anal

gesi

a

cont

inue

s i.v

. mor

phin

e3rd

cho

ice

bupi

vaca

ïne

0.1

25%

+ fe

ntan

yl 2

g/m

L

patie

nt c

ontro

lled

mor

phin

e i.v

.2n

d cho

ice

4mL

bupi

vaca

ine

0.25

% +

epi

neph

rin 1

:200

,000

Th12

-L1

± 1

25m

g bu

piva

caïn

e 0.

25%

+ 1

00μg fe

ntan

yl

rocu

roni

um 0

.6m

g/kg

fent

anyl

1.5

-3μg/

kgpr

opof

ol 2

-2.5

mg/

kgIn

duct

ion

test

-sto

p 6a

m d

ay 3

post

-ope

rativ

e

epid

ural

1st c

hoice

tab

le 1

pre-

med

icat

ion

lora

zepa

m 1

mg

even

ing

prio

r to

surg

ery

para

ceta

mol

100

0mg

lora

zepa

m 1

mg

mor

ning

of s

urge

ry

anes

thes

ia

500m

l BL

500m

l Vol

uven

; 100

0ml B

L 50

0ml

Volu

ven;

150

0ml B

L tra

nsfu

sion

acco

rdin

g to

hem

oglo

bin

Star

t 500

ml V

oluv

esu

bstit

utio

n bl

ood

loss

(BL)

ther

eafte

r 8m

l/kg/

hr1s

t hr 2

0ml/k

gla

ctet

ed ri

nger

’s s

olut

ion

mai

nten

ance

Peri-

oper

ativ

e i.v

. flu

ids

Res

pira

tion

isoflu

rane

.0.8

% e

nd ti

dal

Mai

nten

ance

per

i-ope

rativ

e ca

re a

nd p

ost-o

pera

tive

anal

gesi

a pr

otoc

ol

Page 201: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

200

Gastrointestinal transit studiesGastric emptying was assessed 24hrs after surgery. Patients were asked to drink 100mL

of tap water labeled with 4 MBq 111In-DTPA (Tyco Healthcare, Petten, The Netherlands).

Sixty min after ingestion, a 5-min acquisition was performed in a 128 matrix with the

patient in supine position using a single head gamma camera (Siemens Diacam, Siemens,

Hoffman Estates, Il, USA) fitted with a medium energy collimator. The following formula

was used to calculate the relative gastric content (counts stomach-(pixels stomach*(counts

background/pixels background))* 100 and was depicted as percentage of activity present

in the stomach compared to the total activity in the abdominal region of interest, corrected

for background.

Colon transit was assessed 48 and 72 hrs after surgery. For this, 2 additional 5min-

acquisitions were performed 24 and 48 hrs after ingestion of the radio-labeled water

using the same single head gamma camera and settings also used for gastric emptying.

To enable calculation of colon transit, the colon was subdivided in 7 segments (i.e. 1 =

ascending colon, 2 = right colonic flexure, 3 = transverse colon, 4 = left colonic flexure, 5

= descending colon, 6 = sigmoid / rectum and 7 = stool). The centre of mass model22 was

applied expressing colonic transit as 24 and 48hrs postprandial geometrical centre (GC)

of activity. To correct for the influence of oro-cecal transit the 24hr shift (i.e. delta) in colon

GC was also calculated (GC 48hrs – GC 24hrs). Interpretation and calculation of gastric

retention and colon transit was done by one staff physician (RJB) of the nuclear medicine

department on a Hermes (Nuclear Diagnostics, Sweden) workstation.

Data collectionDuring hospital admission, patients were visited at least once daily, by a trial nurse and/

or research physician, for clinical evaluation (i.e. diet, first passage of flatus, first bowel

movement, vomiting, pain and discomfort). Prior to surgery, most frequently reported

adverse-events for ketotifen known from literature (i.e. drowsiness, dizziness, nausea and

headache)23 were scored daily, using a 100mm visual analog scale (VAS). After surgery,

patients were asked to rate the severity of pain, nausea and abdominal cramping on a VAS

scale every day until discharge. As department policy dictates a minimal hospitalization of

10 days for patients undergoing a radical hysterectomy, duration of hospitalization could

not be used as parameter to evaluate clinical recovery. Instead, patients were deemed

Page 202: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

9M

ast C

ell S

tabi

lizat

ion

in Il

eus:

a R

ando

miz

ed T

rial

201

ready for discharge after tolerance of solid food, occurrence of first bowel movement and

adequate post-surgical pain control with oral analgesics in absence of complications.

Statistical analysisThe pre-defined primary endpoint of efficacy was formulated as the percentage of 111In-

DTPA labeled liquid present in the stomach 1 hr after ingestion, measured 24hrs after

surgery. The secondary endpoints of this study were defined as follows: 1) GC of intra-

colonic mass 24 and 48hrs postprandially and the 24hr colon transit, i.e. delta GC between

24hrs and 48hrs after ingestion of 111In-DTPA labeled water; 2) time until ready for hospital

discharge; 3) time until first flatus in hrs after surgery; 4) time until first bowel movement in

hrs after surgery and 5) degree of post-operative pain, nausea and abdominal cramping

during first 5 days post-operative (mean time until ready for discharge) calculated as area

under the curve. As this study was designed as a proof of principle study, per-protocol

analysis was applied on all data.

Previous studies on gastrointestinal transit in healthy females24 indicated that 16 patients

would suffice to identify a >10% significant (p<0.05) difference in gastric retention 1hr after

ingestion of a non-caloric liquid test meal between placebo and ketotifen treated patients,

providing a 90% power.

Data were non-parametrically distributed and therefore expressed as median values and

inter-quartile range. For paired and unpaired data the Wilcoxon signed rank or the Mann-

Whitney U test was used respectively. To identify potential confounders in ordinal data

sets a Chi-square test was applied. For analyses of clinical symptom VASscores, the area

under the curve (AUC) was calculated for each individual patient. These AUC values were

statistically tested using an independent sample test. P<0.05 was considered statistically

significant. Statistical analysis was performed using SPSS 12.02 software for Windows

(SPSS Inc. Chicago, Ill, USA).

Page 203: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

202

ResultsStudy subjectsBetween June 2004 and March 2006 a total of 60 female patients were enrolled in this

study, 20 patients in each treatment group (i.e. placebo, 4mg ketotifen and 12mg ketotifen

per day). One patient, randomized for placebo, withdrew from the trial because of non-drug

related personal reasons. A protocol violation was reported in 15 cases, 3 in the placebo,

7 in the 4 mg and 5 in the 12mg of ketotifen group. Therefore data from 44 patients was

available for full analysis. Six cases were excluded on post-operative day 2 or 3 because

of laxative/prokinetic drug administration (placebo n=1, ketotifen 4mg n=3 and 12mg n=2).

Hence, only gastric emptying studies of these patients were included in the analysis (n=50;

fig.1).

The majority of patients (36) underwent a radical hysterectomy, 14 patients underwent

tumor debulking and 10 patients an explorative laparotomy because of suspicion of

malignant disease. The distribution of the three types of surgery was statistically equal in

all three treatment groups. From the remaining baseline characteristics only ASA-health

classification was not equally distributed (table 2). Pain medication (i.e. paracetamol,

NSAIDs and tramadol) consumption, calculated as median AUC for daily consumption until

ready for discharge, did not differ between the 3 treatment groups (table 2).

Evaluation of adverse events potentially related to ketotifen use revealed no serious

adverse events. None of the patients indicated they were considering withdrawal from the

trial because of side effects. In accordance with previous reports on ketotifen drowsiness

was the most frequently noted adverse event (median VAS for placebo: 0.0 (InterQuartile

Range (IQR) 0.0-0.0 vs. ketotifen 4mg: 0.6 (IQR 0.0-5.5), p=0.02 and 12mg: 2.5 (IQR 0.0-

7.1), p=0.002).

Page 204: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

9M

ast C

ell S

tabi

lizat

ion

in Il

eus:

a R

ando

miz

ed T

rial

203

Enro

lled

n=60

plac

ebo

n=20

keto

tifen

4m

gn=

20ke

totif

en 1

2mg

n=20

com

plet

ed g

astri

c em

ptyin

gn=

17

com

plet

ed s

tudy

n=16

prot

ocol

vio

latio

nla

xativ

es/p

rokin

etics

(n=1

)

prot

ocol

vio

latio

nan

ti-hi

stam

ines

(n=2

)w

ithdr

ew in

form

ed c

onse

nt (n

=1)

com

plet

ed s

tudy

n=13

com

plet

ed g

astri

c em

ptyin

gn=

16

prot

ocol

vio

latio

nm

issed

med

icatio

n (n

=2)

colo

stom

y (n

=1)

anti-

infla

mm

ator

y ag

ents

(n=1

)

prot

ocol

vio

latio

nla

xativ

es/p

rokin

etics

(n=3

)

com

plet

ed s

tudy

n=15

com

plet

ed g

astri

c em

ptyin

gn=

17

prot

ocol

vio

latio

nm

issed

med

icatio

n (n

=1)

surg

ery

canc

eled

(n=1

)an

ti-in

flam

mat

ory

agen

ts (n

=1)

prot

ocol

vio

latio

nla

xativ

es/p

rokin

etics

(n=2

)

Figure 1Flowchart of patient enrolment, protocol violation, exclusion and number of patients completing the study.

Page 205: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

204

ns28

08 (2

646-

2951

)mg

2792

(272

5-29

19)m

g28

75 (2

716-

2893

)mg

pa

race

tam

ol (m

edia

n AU

C)

ns0

(0-0

)mg

0 (0

-0)m

g0

(0-1

25)m

g

NSA

IDs

nsnsns

p=0.

02

nsns

p-va

lue

patie

nt c

hara

cter

istic

s

tabl

e 2

exa

ct v

alue

, mea

n ±

SD o

r med

ian

(inte

r-qua

rtile

rang

e)

a=e

xclu

ded

prio

r to

gast

ric e

mpt

ying

stud

ies

and

b=

afte

r gas

tric

empt

ying

stud

ies

12

1 m

orph

ine

cont

inue

s i.v

.

100

(0-2

38)m

g12

5 (0

-213

)mg

75 (0

-200

)mg

tra

mad

ol

23

4 m

orph

ine

patie

nt c

ontro

lled

i.v.

1615

15 e

pdur

al

anal

gesi

a

3.8

± 1.

73.

3 ±

1.6

SD4.

7 ±

1.8

dura

tion

of s

urge

ry (h

rs)

2 (1

-2)

2 (2

-2)

1 (1

-2)

ASA-

PS s

core

(sca

le 1

-5)

45

1 e

xplo

rativ

e la

paro

tom

y3

83

deb

ulki

ng

137

16 r

adic

al h

yste

rect

omy

type

of s

urge

ry

48 ±

10

55 ±

11

47 ±

12

age

(yrs

)

n=20

n=20

n=20

enro

llmen

t

keto

tifen

12m

gke

totif

en 4

mg

plac

ebo

para

met

er

Page 206: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

9M

ast C

ell S

tabi

lizat

ion

in Il

eus:

a R

ando

miz

ed T

rial

205

placebo ketotifen4mg

ketotifen12mg

0102030405060708090

100

p=0.01

rela

tive

gast

ric c

onte

nts

(%)

Figure 2Scintigraphical evaluation of gastric emptying of an 111In labeled non-caloric liquid test-meal 24hrs after surgery. Gastric retention determined 1hr postprandial, depicted as median percentage in stom-ach compared to total abdominal region corrected for background.

Gastrointestinal transit1. Gastric emptying

Gastric emptying was determined 24 hrs after surgery. One hour after ingestion of 100 mL

of radio-labeled tap water, the residual gastric radioactivity was calculated as measure of

emptying. As shown in figure 2, gastric emptying of patients treated with placebo varied

considerable, ranging from complete emptying to gastric stasis with more than 90% of

radiolabeled material still present in the stomach. The median gastric retention was 16 %

(IQR 5-75). Treatment with 4 mg ketotifen did not significantly change gastric emptying

(gastric retention: 18 % (IQR 3-45), p=0.6) compared to placebo. In contrast, gastric

emptying of patients treated with 12mg ketotifen was significantly improved with a median

gastric retention of 3% (IQR 1-7), p=0.01) (fig. 2).

Page 207: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

206

Fifteen of the 17 patients had almost completely emptied their stomach one hour after

ingestion of the radiolabeled water. To put things in perspective, 47% of patients in the

placebo group and 44% in 4mg ketotifen showed >20% residual gastric content one hour

after ingestion of radiolabeled tab water in comparison to only 12% of patients in the 12mg

ketotifen.

A placebo

GC24h postprandial

GC48h postprandial

0

1

2

3

4

5

6

colo

n se

gmen

t

Bcebo

postprandial po tprandial

2

3

4

5

ketotifen 4mg

GC24h postprandial

GC48h postprandial

0

1

2

3

4

5

6

colo

n se

gmen

t

A

C

0

1

2

4

c

l

e

ketotifen 12mg

GC24h postprandial

GC48h postprandial

0

1

2

3

4

5

6

colo

n se

gmen

t

placebo ketotifen4mg

ketotifen12mg

-1

0

1

2

3p=0.07

24 h

our c

olon

tran

sit

cn

gn

0

5

colo

n se

g

GC

GC

o

mnt

D

Figure 3Scintigraphical evaluation of intestinal transit of an 111In labeled non-caloric liquid testmeal 24hrs after surgery. a) colonic transit time, depicted as median shift of geometrical center (GC) of colonic contents in number of (predefined) segments per 24hrs; b) individual GC’s of colon transit 24 and c) 48hrs postprandial for each consecutive treatment group. Note the smaller distribution in the ketotifen treated group 24hrs postprandial, not present at 48hrs postprandial. d) 24 to 48hr GC-shift calculated to correct for potential study drug related influences on (small) intestinal motility. Dottedline indicates median.

Page 208: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

9M

ast C

ell S

tabi

lizat

ion

in Il

eus:

a R

ando

miz

ed T

rial

207

2. Colonic transit

Colonic transit was determined 48 and 72 hrs after surgery, or 24 and 48 hrs after ingestion

of the radio-labeled tap water. As depicted in figure 3, colonic transit varied significantly

in patients treated with placebo; 24 hrs after intake the GC was still located in the small

intestine in one patient but had already moved towards the left colonic flexure in others. The

median GC was 1.9 (1.0-2.8). A similar distribution was observed in the group of patients

treated with 4 mg ketotifen. The median GC at t=24hrs after intake was 1.2 (1.0-2.4) (NS

compared to placebo). In contrast to placebo and 4 mg, the GC of patients treated with 12

mg ketotifen was less dispersed and varied mainly (except in one patient) between 1 and

2.5, with a median of 1.5 (1.3-2.4) (NS compared to placebo). These data indicate that most

of the radiolabeled material was located in the right colon. In this respect, it is important to

notice that patients were still on medication at this point, which was only discontinued at

the end of the day.

The next day, patients were off medication when the colonic transit was assessed at t=48hrs

after intake. In patients treated placebo, the GC had shifted more distally with 0.8 (0.0-1.1)

unit towards 2.5(1.9- 4.0). The calculated GC of activity did not show a statistical difference

between the placebo and the two doses of ketotifen (fig.3a-c).

Based on the observation that the GC at t=24hrs in patients treated with 12 mg ketotifen

tended to be located more proximally, despite the improvement of gastric emptying, we

hypothesized that the highest dose of ketotifen might delay intestinal/colonic transit. To

eliminate this possible confounding effect, we calculated the shift in GC between t=24hrs

and t=48hrs. In the placebo treated group this GC shift over 24hrs was 0.8 (0.0-1.1)

segments compared to 0.6 (0.0-1.2) and 1.2 (0.2-1.4) segments in the ketotifen 4 and

12mg treated groups respectively, showing a trend towards significance for the 12 mg

group (p=0.07 placebo vs. ketotifen 12mg) (fig.3d).

Page 209: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

208

Clinical evaluationTable 3 depicts the outcome of clinical endpoints and marks the time interval between

the end of the surgical procedure and the occurrence of the indicated event. None of the

clinical endpoints of gastrointestinal recovery were significantly improved after surgery.

VAS scores for pain, nausea or abdominal cramping are plotted in fig. 4. The area under the

curves (AUC) were calculated showing a significant improvement for abdominal cramping

in the 12mg dose (median AUC for placebo 10.4 (3.2-18-9); ketotifen 4mg 6.4 (0.0-13.0),

p= ns; ketotifen 12mg 4.6 (0.5-8.4), p= 0.03) but not for pain (AUC for placebo 6.9 (3.3-

10.6) vs. ketotifen 4mg 8.0 (7.2-12.5), p= ns and 12mg (2.4 (0.0-18.8), p= ns) and nausea

(AUC for placebo 3.0 (0.6-6.9) vs. ketotifen 4mg 7.0 (0.2-15.0), p= ns and 12mg 1.7 (0.0-

13.0), p= ns).

abdominal cramping

0 1 2 3 4 50123456789

10

day post-surgery

pain

0 1 2 3 4 50123456789

10

day post-surgery

p=0.03

placeboketotifen 4mgketotifen 12mg

placeboketotifen 4mgketotifen 12mg

placeboketotifen 4mgketotifen 12mg

ns

nausea

0 1 2 3 4 50123456789

10

day post-surgery

VAS-

scor

e

VAS-

scor

e

VAS-

scor

e

ns

Figure 4Clinical evaluation of pain, nausea and abdomi-nal cramping over the first 5 postoperative days (i.e. median time till ready for discharge). Note that median time of epidural/i.v. analgesia is 3 days.

Page 210: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

9M

ast C

ell S

tabi

lizat

ion

in Il

eus:

a R

ando

miz

ed T

rial

209

clin

ical

(sec

onda

ry) e

ndpo

ints

p-va

lue

vs. p

lace

bo

nsnsnsns

p-va

lue

vs. p

lace

bo

nsnsnsns

med

ian

time

till e

vent

occ

urre

d in

hrs

afte

r sur

gery

(int

er-q

uarti

le ra

nge)

tabl

e 3

119

(96-

130)

119

(105

-147

)11

9 (9

4-12

5)tim

e un

til re

ady

for h

ospi

tal

disc

harg

e

72 (7

2-96

)96

(72-

132)

96 (7

2-10

8)tim

e un

til s

olid

food

inta

ke11

4 (9

8-12

5)11

8 (1

08-1

20)

114

(91-

125)

time

until

firs

t bow

el m

ovem

ent

30 (1

9-51

)43

(20-

74)

23 (2

0-36

)tim

e un

til fi

rst f

latu

s

keto

tifen

12m

gke

totif

en 4

mg

plac

ebo

endp

oint

Page 211: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

210

DiscussionPostoperative ileus is an iatrogenic disorder characterized by impaired and disturbed

motility of the entire gastrointestinal tract. Spontaneous recovery of intestinal transit or

coordinated motility is initiated first in the small intestine, approximately 24hrs after surgery,

but it may last up to 3 to 5 days before gastric and colonic function have returned to normal25.

One approach to enhance this process is stimulation of gastrointestinal motility with

potent prokinetics, of which cisapride is the most studied drug. Cisapride administered i.v.

induced a significantly faster propagation of radiopaque markers in the colon accompanied

by a significantly earlier first bowel movement in patients undergoing cholecystectomy26.

Most studies however fail to demonstrate clinical improvement15. Treatment with 3 times

30 mg rectal cisapride induced some changes in motor activity but did not enhance the

recovery to normal motility or clinical outcome in patients who underwent major intra-

abdominal surgery27. Similarly, Hallerback et al. failed to demonstrate changes in time to

first bowel movement after upper gastrointestinal or colonic surgery by rectal administration

of cisapride10. One might argue that the absence or moderate effect of prokinetics could

result from the fact that the underlying cause of postoperative ileus, i.e. increased inhibitory

neural input to the gastrointestinal tract, has not been targeted. Especially as recent findings

indicate that inflammation induced by handling of the intestine continuously drives this

inhibitory input16, 28, prevention of this inflammatory response could embody an alternative

therapeutic approach.

Previously, we demonstrated that mast cells play an important role in the development

of the inflammatory response to intestinal handling20. Animals lacking mast cells do not

develop intestinal inflammation after surgery, whereas treatment with mast cell stabilizers

block the occurrence of handling-induced inflammation in wild type animals. Conversely,

mast cell degranulation with compound 48/80 induces a local inflammatory response in the

exposed intestinal loop inducing delayed gastric emptying20. In line with these animal data,

we recently showed in man that a conventional open hysterectomy, but not a laparoscopic

adnexectomy or transvaginal hysterectomy results in the release of the mast cell mediator

tryptase in peritoneal lavage fluid and triggers the influx of leukocytes in the intestinal

muscularis18. Although there is abundant evidence in animals that prevention of surgery-

Page 212: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

9M

ast C

ell S

tabi

lizat

ion

in Il

eus:

a R

ando

miz

ed T

rial

211

induced intestinal inflammation shortens postoperative ileus and is an effective alternative

treatment, human studies supporting this principle are still lacking. Therefore, we designed

the current pilot study investigating the effect of ketotifen, a mast cell stabilizer used in

allergic disorders such as hay fever, on postoperative gastric emptying of liquids. This

parameter was chosen as primary outcome parameter as it parallels our animal model, and

is a reliable and reproducible read-out of gastric motility with accepted clinical relevance.

Patients ingested 100 mL of radiolabeled tap water 24 hrs after the surgical procedure and

gastric retention was determined by scintigraphic imaging one hr later. In patients treated

with placebo or 4 mg ketotifen, gastric emptying varied considerable: some patients had

emptied their stomach almost completely whereas others had a severe gastric stasis with

more than 90% of the radiolabel still present in the stomach. In contrast, gastric emptying

of patients treated with 12 mg ketotifen was significantly faster with almost complete

emptying in 15 of the 17 patients. These findings show that mast cell stabilization restores

gastric emptying after abdominal surgery and provide indirect support for the concept that

intervention with the mast cell – inflammation cascade may represent a new therapeutic

approach for postoperative ileus. It should be emphasized though that a direct prokinetic

effect of ketotifen can not be excluded, especially as no human data on the effect of

ketotifen or mast cell stabilizers on gastric emptying are available. In rats however, the

mast cell stabilizers disodium cromoglycate and FPL-52694 significantly inhibited gastric

motor activity indirectly arguing against this possibility29.

In addition to gastric emptying, we also monitored clinical parameters and colonic transit,

measured by means of the GC 24 and 48 hrs after ingestion of radiolabeled tab water,

as secondary outcome parameter of this pilot study. No significant effect of ketotifen was

detected on colonic transit. As shown in figure 2, the variation in colonic transit in the placebo

group was very large, implying that this negative finding might represent a type II error. In

fact, the same applies for the effect on symptoms and clinical recovery. Only abdominal

cramping was reduced by ketotifen 12 mg, whereas nausea, vomiting, pain and clinical

recovery parameters remained unaltered. Alternatively, animal data indicate that ketotifen

inhibits colonic motility possibly obscuring the beneficial effect of interference with the mast

cell-inflammation cascade. Ketotifen indeed has mild anti-cholinergic properties23, but also

relaxes the mouse colon and inhibits small intestinal contractions evoked by carbachol and

nerve stimulation30. In this respect, it should be emphasized that patients were still treated

Page 213: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

212

with ketotifen when colonic transit at t=24 hrs was determined. The observation that the GC

of all but one patient treated with 12 mg ketotifen was situated in the right colon at t=24 hrs,

whereas it had moved up to the left colonic flexure in some patients treated with placebo,

indirectly supports this possibility. Moreover, when ketotifen was stopped at the end of

postoperative day 2, the transit of the GC between t=24 and t=48 hrs indeed tended to be

faster in the ketotifen group compared to the placebo group. These considerations would

imply that ketotifen treatment in future studies must be stopped immediately after surgery,

similar to the treatment regimen used in our animal study20.

Although this study clearly opens perspective for future treatment of postoperative ileus,

there are some drawbacks that need to be considered. First, there were a relatively large

number of dropouts, mainly due to protocol violation in the initial phase of the study. This

was caused by the administration of drugs on the ward that were not allowed according

to the study protocol, defined as exclusion criteria. However, the number of dropouts is

comparable in all three patient groups making it rather unlikely that this will affect the

outcome of the study. Second, a large variation was observed in gastric emptying and

colon transit. Especially gastric emptying for liquids was almost completed in a significant

proportion of patients, even after placebo treatment. Emptying of a solid caloric test-

meal would have been more appropriate and might have yielded more consistent results,

perhaps showing an even greater difference between the treatment arms. However as

data on early postoperative gastric emptying in patients are lacking, manly for safety

reasons, emptying of liquids was evaluated in stead. A third point is the lack of baseline

gastrointestinal transit measurements. This design would have allowed comparison before

and after surgery in each individual patient, reducing variability and increasing the power of

the study. Nevertheless, even with the current design our study showed a dose-dependent

effect of ketotifen on gastric emptying.

Our study may be of great clinical relevance, as it partly confirms our animal data

demonstrating that mast cell stabilization restores surgery-induced delayed gastric

emptying. Based on our animal research data and our previous findings in patients, this

effect most likely result’s from blockade of the inhibitory neural input to the stomach driven

by intestinal inflammation. If this concept indeed proves to be important in the pathogenesis

of postoperative ileus in human, the treatment of this iatrogenic disorder will change

Page 214: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

9M

ast C

ell S

tabi

lizat

ion

in Il

eus:

a R

ando

miz

ed T

rial

213

dramatically in the near future. There are however important issues that still need to be

addressed or require improvement. It remains to be studied if the observed effect on gastric

emptying indeed results from blockade of intestinal inflammation. Secondly, future studies

are required to demonstrate whether results can be further improved, i.e. improvement of

colonic transit and clinical recovery. This may be achieved by changing the concentration

or route of administration for ketotifen. Higher dosages injected intravenously before and

during surgery or even lavage of the abdominal cavity with ketotifen could be alternative

treatment protocols, avoiding the possible inhibitory effect of ketotifen on gastrointestinal

motility. Nevertheless, we feel that our observation is an important step forward encouraging

larger clinical studies with ketotifen or other more potent mast cell stabilizers.

Page 215: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

214

Reference ListCollins TC, Daley J, Henderson WH, Khuri SF. Risk factors for prolonged length of stay after 1. major elective surgery. AnnSurg 1999;230(2):251-9.Prasad M, Matthews JB. Deflating postoperative ileus. Gastroenterology 1999;117(2):489-92.2. Boeckxstaens GE, Hirsch DP, Kodde A, et al. Activation of an adrenergic and vagally-medi-3. ated NANC pathway in surgery-induced fundic relaxation in the rat. NeurogastroenterolMotil 1999;11(6):467-74.De Winter BY, Boeckxstaens GE, De Man JG, Moreels TG, Herman AG, Pelckmans PA. 4. Effect of adrenergic and nitrergic blockade on experimental ileus in rats. BrJPharmacol 1997;120(3):464-8.De Winter BY, Boeckxstaens GE, De Man JG, et al. Effect of different prokinetic agents and a 5. novel enterokinetic agent on postoperative ileus in rats. Gut 1999;45(5):713-8.Seta ML, Kale-Pradhan PB. Efficacy of metoclopramide in postoperative ileus after exploratory 6. laparotomy. Pharmacotherapy 2001;21(10):1181-6. Cheape JD, Wexner SD, James K, Jagelman DG. Does metoclopramide reduce the 7. length of ileus after colorectal surgery? A prospective randomized trial. DisColon Rectum 1991;34(6):437-41.Jepsen S, Klaerke A, Nielsen PH, Simonsen O. Negative effect of Metoclopramide in 8. postoperative adynamic ileus. A prospective, randomized, double blind study. BrJSurg 1986;73(4):290-1.Brown TA, McDonald J, Williard W. A prospective, randomized, double-blinded, placebo-con-9. trolled trial of cisapride after colorectal surgery. AmJSurg 1999;177(5):399-401.Hallerback B, Bergman B, Bong H, et al. Cisapride in the treatment of postoperative ileus. 10. AlimentPharmacolTher 1991;5(5):503-11.Boghaert A, Haesaert G, Mourisse P, Verlinden M. Placebo-controlled trial of cisapride in post-11. operative ileus. Acta AnaesthesiolBelg 1987;38(3):195-9. Smith AJ, Nissan A, Lanouette NM, et al. Prokinetic effect of erythromycin after colorectal sur-12. gery: randomized, placebo-controlled, double-blind study. DisColon Rectum 2000;43(3):333-7.Bonacini M, Quiason S, Reynolds M, Gaddis M, Pemberton B, Smith O. Effect of intravenous 13. erythromycin on postoperative ileus. AmJGastroenterol 1993;88(2):208-11.Bungard TJ, Kale-Pradhan PB. Prokinetic agents for the treatment of postoperative ileus in 14. adults: a review of the literature. Pharmacotherapy 1999;19(4):416-23. Holte K, Kehlet H. Postoperative ileus: a preventable event. BrJSurg 2000;87(11):1480-93.15. de Jonge WJ, van den Wijngaard RM, The FO, et al. Postoperative ileus is maintained by 16. intestinal immune infiltrates that activate inhibitory neural pathways in mice. Gastroenterology 2003;125(4):1137-47.The FO, de Jonge WJ, Bennink RJ, van den Wijngaard RM, Boeckxstaens GE. The ICAM-1 17. antisense oligonucleotide ISIS-3082 prevents the development of postoperative ileus in mice. BrJPharmacol 2005.The FO, Bennink RJ, Ankum WM, et al. Intestinal handling induced mast cell activation and 18. inflammation in human post-operative ileus. Gut 2007.Kalff JC, Turler A, Schwarz NT, et al. Intra-abdominal activation of a local inflammatory re-19. sponse within the human muscularis externa during laparotomy. AnnSurg 2003;237(3):301-15.de Jonge WJ, The FO, van der CD, et al. Mast cell degranulation during abdominal surgery 20. initiates postoperative ileus in mice. Gastroenterology 2004;127(2):535-45.Saklad M. Grading of patients for surgical procedures. Anesthesiology 1941;2:4. 21. Kamm MA. The small intestine and colon: scintigraphic quantitation of motility in health and 22. disease. EurJNuclMed 1992;19(10):902-12.Grant SM, Goa KL, Fitton A, Sorkin EM. Ketotifen. A review of its pharmacodynamic and 23. pharmacokinetic properties, and therapeutic use in asthma and allergic disorders. Drugs 1990;40(3):412-48.

Page 216: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

9M

ast C

ell S

tabi

lizat

ion

in Il

eus:

a R

ando

miz

ed T

rial

215

Bennink R, Peeters M, Van dMV, et al. Evaluation of small-bowel transit for solid and 24. liquid test meal in healthy men and women. EurJNuclMed 1999;26(12):1560-6.Miedema BW, Johnson JO. Methods for decreasing postoperative gut dysmotility. Lan-25. cet Oncol 2003;4(6):365-72.Tollesson PO, Cassuto J, Rimback G, Faxen A, Bergman L, Mattsson E. Treatment of 26. postoperative paralytic ileus with cisapride. Scandinavian journal of gastroenterology 1991;26(5):477-82.Benson MJ, Roberts JP, Wingate DL, et al. Small bowel motility following major intra-27. abdominal surgery: the effects of opiates and rectal cisapride. Gastroenterology 1994;106(4):924-36.Kalff JC, Carlos TM, Schraut WH, Billiar TR, Simmons RL, Bauer AJ. Surgically induced 28. leukocytic infiltrates within the rat intestinal muscularis mediate postoperative ileus. Gastroenterology 1999;117(2):378-87.Takeuchi K, Nishiwaki H, Okabe S. Cytoprotective action of mast cell stabilizers 29. against ethanol-induced gastric lesions in rats. Japanese journal of pharmacology 1986;42(2):297-307. Abu-Dalu R, Zhang JM, Hanani M. The actions of ketotifen on intestinal smooth 30. muscles. EurJPharmacol 1996;309(2):189-93.

Page 217: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

10

Page 218: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

10C

hapt

er 1

0Summary and conclusions

Page 219: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

218

MSummary and conclusionsMore than a millennium after its first written documentation, postoperative ileus still is a

prevalent clinical condition with significant morbidity and socio-economical impact1, 2. Even

to date, every patient undergoing abdominal surgery remains hospitalized for several

days as he or she will experience a period of nausea, lack of appetite and inability to eat

or defecate3. Both studies in animal models4-7 and in man1, 8 reveal that these symptoms

result from absence or disturbed gastrointestinal motility and failed propulsion of intestinal

contents, known as postoperative ileus. Mainly pain stimuli during surgery, induced by

skin incision, opening of the peritoneum, but above all handling of the intestine, have

been identified as a major cause of this instantaneous “paralysis” of the intestines4, 9, 10.

Until recently, it was generally accepted that activation of inhibitory neural pathways by

nociceptive / mechanical stimuli explained the generalized impairment of gastrointestinal

motility. Pharmacological neural blockade, section of nerves or the spinal cord, afferent

nerve ablation with capsaicin and identification of activated nerve pathways and brain

nuclei all confirmed this hypothesis4, 5, 7, 11, 12. Most experiments however were performed

during surgery or evaluated gastrointestinal function within a time frame of up to 3 hours

after surgery. By now, we know that this period reflects the first early phase of postoperative

ileus and only represents the tip of the iceberg. In this thesis, we indeed describe that this

early phase is followed by a second prolonged phase triggered by inflammation of the

handled intestine.

During each surgical procedure in the peritoneal cavity, the intestines will have to be replaced

from their original location, either to reach the organ of interest, to inspect the intestine for

abnormalities or to isolate the diseased segment for resection. Although the exact initial

trigger remained unclear, it became clear that the extent of intestinal handling might be

one of the major determinants of the severity of postoperative ileus. This observation

undoubtedly has been an important stimulus for the development and the introduction of

minimal invasive surgery, associated with a significant reduction in postoperative ileus and

duration of hospitalization13-15. It was not until a few years ago that we began to understand

how intestinal handling could lead to prolonged inhibition of intestinal neuromuscular

function. Kalff and coworkers demonstrated that 3 to 4 hours after mechanical manipulation

of the intestine, the muscularis became infiltrated by inflammatory cells16. In rodents, this

Page 220: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

10

Sum

mer

y an

d co

nclu

sions

219

phenomenon led to impaired transit of intestinal content and reduced in vitro contractile

activity of inflamed muscle strips for more than 24 hours17, this thesis. This finding has led

to the “inflammatory” hypothesis suggesting that the prolonged duration of postoperative

ileus rather results from inflammation-induced impairment of gastrointestinal motility,

and not from activation of inhibitory neural pathways. Based on these observations, the

pathophysiology of postoperative ileus is now subdivided in 2 phases; an instantaneous

short lasting neurogenic phase resulting from activation of nociceptive neural pathways

during surgery, and a second late-onset (after 3-4 hours) inflammatory phase. Given its

duration, the second phase is obviously the most important one, at least from a clinical

point of view. In the current thesis, we have been focusing on this second phase and

have tried to unravel the cells and mechanisms involved in its pathophysiology in order to

develop more efficient therapeutic strategies to shorten postoperative ileus.

Although the inflammatory theory certainly explains the prolonged nature of postoperative

ileus or in other words, explains how gastrointestinal motility remains disrupted even

though the initial surgical stimulus has ceased, it fails to explain the generalized nature

of postoperative ileus. Indeed, if we accept that intestinal handling leads to inflammation

of the handled segment, then how does this explain dysfunction of those areas of the

intestine that have not been handled during surgery? One explanation could be that surgery

triggers a systemic inflammatory response via for instance the release of pro-inflammatory

mediators / cytokines in the systemic circulation. However, 24 hours after manipulation of

the small intestine, we observed inflammation of the manipulated segments, but not in other,

non-handled, areas of the gastrointestinal tract. To investigate the mechanisms leading to

the generalized impairment of gastrointestinal motility, we developed a mouse model in

which gastric emptying was used a read-out to determine the degree of postoperative ileus

(chapter 2). The small intestine was gently manipulated during 5 minutes after which the

abdomen was closed and the animals were allowed to recover. Twenty-four hours later,

a radio-labeled meal was gavaged and gastric emptying was determined. Only animals

that underwent intestinal manipulation during a laparotomy revealed delayed gastric

emptying, whereas those that underwent a laparotomy only had normal transit (chapter 2). Similarly, inflammation of the muscularis was only observed in animals that underwent

abdominal surgery (intestinal manipulation) (chapter 2). Most importantly, the inflammation

was limited to the handled region, i.e. the small intestine, but was absent in the stomach.

Page 221: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

220

Nevertheless, gastric emptying was significantly delayed in our model up to 48 hours after

surgery (chapter 2). Moreover, we showed that prevention of influx of inflammatory cells by

several interventions, like antibodies or antisense olignucleotides to the adhesion molecule

ICAM-1, restored gastric emptying, indicating that this infiltrate was indeed responsible for

the observed ileus (chapter 3).

How then can local inflammation of the small intestine lead to delayed gastric emptying?

We hypothesized that activation of neural pathways by the infiltrate must be involved. To

confirm this concept, animals were pretreated with the ganglion blockers guanethidine

and hexamethonium to inhibit neurotransmission (chapter 2). These experiments

indeed showed that gastric emptying was normalized by these agents, even though the

inflammation in the small intestine was still present. To further prove that the local infiltrate

activated inhibitory neural pathways, we stained the spinal cord for c-fos expression, a

marker of neural activation (chapter 2). Animals that underwent abdominal surgery, but not

those subjected to a laparotomy only, showed c-fos expression in the spinal cord. When

manipulation-induced inflammation was blocked by pretreatment with adhesion molecule

neutralizing antibodies, the increase in c-fos expression was prevented, clearly confirming

our hypothesis (chapter 2). From these experiments, we concluded that the prolonged late

phase of postoperative ileus results from neuro-immune interaction between inflammatory

cells in the manipulated segment and its afferent innervation. This interaction activates

an inhibitory adrenergic neural pathway synapsing in the spinal cord, affecting the entire

gastrointestinal tract.

The next crucial question that arose was how handling of the intestine triggers the influx of

inflammatory cells. Obviously, tissue damage will attract immune cells and will contribute to

the local inflammatory process. However, we reasoned that intense activation of nociceptive

nerve fibers would play a more important role, especially as earlier studies showed that

intense activation of afferent nerve fibers is an important trigger for local inflammation, also

referred to as neurogenic inflammation18, 19. Afferent nerve fibers, when intensely activated,

release neuropeptides like Calcitonine Gene Related Peptide (CGRP) and substance P

at the site of stimulation19. These peptides are potent pro-inflammatory mediators, mainly

by their capacity to stimulate mast cells18, 20. Mediators released by mast cells will not only

directly attract inflammatory cells, but will also lead to the transient increase in mucosal

Page 222: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

10

Sum

mer

y an

d co

nclu

sions

221

permeability21 and bacterial translocation previously described after intestinal handling22.

Schwarz et al. indeed elegantly demonstrated that intestinal manipulation leads to influx

of intraluminal micro-spheres during a time window of 4 hours after manipulation22. This

transient disruption of the intestinal barrier allows intraluminal bacteria to enter the intestinal

wall, an important trigger to activate the immune system. Activation of resident macrophages,

located in between the longitudinal and circular muscle layer23, has been described to

occur a few hours after intestinal manipulation, as shown by upregulation of IL-6, iNOS

and LFA-117, 24, 25. In chapter 7, we confirmed the important role of mast cells and showed

that manipulation-induced inflammation and ileus were reduced in animals pre-treated with

mast cell stabilizers and in animals lacking mast cells (W/Wv mice). Reconstitution of mast

cells in W/Wv mice restored the capacity to develop an inflammatory response following

intestinal manipulation. Also in man, we demonstrated the release of mast cell mediators

in the peritoneal cavity, even after gentle inspection of the intestinal at the beginning of

the surgical procedure (chapter 8). In line with our animal findings, mast cell activation

was followed by the upregulation and release of inflammatory mediators such as IL-6,

IL-8, iNOS and ICAM-1. This process ultimately led to the influx of inflammatory cells in

to the muscularis propria of the resected intestinal tissue specimen at the end of surgery.

Interestingly, this cascade of events occurred almost exclusively in patients who underwent

conventional (open) surgery, but not in patients who underwent minimal invasive surgery.

Using radio-labelled leukocyte SPECT scanning, an increase in influx of leukocytes 24

hr after surgery (compared to baseline pre-operative scanning) was only demonstrated

after open, but not after laparoscopic abdominal surgery in patients (chapter 8). These

findings demonstrate both in mice and man that mast cell activation triggered via surgical

bowel manipulation represents an important initial step in the cascade of events leading to

intestinal inflammation and postoperative ileus.

As shown in the Summarizing Figure, increased permeability induced by mast cell activation

leads to bacterial translocation, most likely contributing to the activation of resident

macrophages after intestinal handling, described earlier by Kalff et al.24. Interestingly,

Borovikova et al. reported dampening of macrophage activation by the vagus nerve26. In

a model of sepsis, these investigators demonstrated increased survival and improvement

of blood pressure after LPS infusion when the vagus nerve was electrically stimulated.

Acetylcholine, released by the vagus nerve, was shown to interact with alpha7 nicotinic

Page 223: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

222

Page 224: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

10

Sum

mer

y an

d co

nclu

sions

223

Summarizing figure: (see fullcolor chapter 11)Summary of the pathophysiology of postoperative ileus. The inevitable handling of the intestines dur-ing abdominal surgery (A) results in the instant activation and degranulation of mast cells (B). The latter leads to transient intestinal barrier dysfunction enabling luminal bacteria to enter the intestinal wall (C). A network of macrophages residing between the circular and longitudinal muscle layer prob-able phagocytize these bacteria and become activated (D). These events result in upregulation of the adhesion molecules ICAM-1 and LFA-1 and recruitment of leukocytes from the circulation in to the intestinal muscle layer (E). This local inflammation then activates inhibitory neural pathways explain-ing the sustained general inhibition of gastrointestinal motility during postoperative ileus (F). This cascade identifies several new targets for therapeutic strategies to shorten or prevent postoperative ileus (indicated in rectangles on right).

receptors on macrophages, resulting in a reduction in the release of the pro-inflammatory

cytokines TNF-alpha27. Especially as the gastrointestinal tract is largely under control of

the vagus nerve, we investigated whether electrical nerve stimulation could also intervene

with the activation of the resident macrophages thereby reducing the inflammatory

response and ileus following abdominal surgery (chapter 4). Indeed, electrical vagus

nerve stimulation diminished intra-peritoneal release of TNF, MIP-2 and IL-6 three hrs after

surgery in our ileus model, indicating a reduction of the activation of macrophages during

surgery. Accordingly, the number of leukocytes recruited to the intestinal muscle layer was

significantly reduced 24 hrs later, associated with a normal gastric emptying rate (chapter

4). To further confirm the anti-inflammatory properties of the vagus nerve, experiments

were performed with CNI 1493, a p38 MAPKinase inhibitor shown to reduce inflammatory

responses in a vagus nerve dependent manner when injected i.c.v.28, 29. Like electrical

nerve stimulation, this intervention reduced inflammation and restored gastric emptying,

an effect abolished by vagotomy (chapter 6). This set of experiments indicates that also

in the gastrointestinal tract, the vagus nerve exerts an important anti-inflammatory input

contributing to the control of the innate immune response. To elucidate how acetylcholine

exerts its anti-inflammatory effect on macrophages, peritoneal macrophages were isolated

and activated in vitro with LPS in the presents of nicotine. This agonist indeed reduced the

release of TNF, IL-6 and MIP-2 via its alpha7 acetylcholine receptor subtype (chapter 4).

We identified the signal transduction pathway mediating the inhibitory effect of nicotine and

demonstrated that activation of the alpha7 receptor subtype on macrophages results in the

subsequent phosphorylation of Jak2 and STAT3 decreasing the release of inflammatory

mediators (chapter 4). The importance of this signaling cascade is illustrated by the fact

that manipulation induced inflammation cannot be reduced through vagus nerve stimulation

in STAT3 conditional knock-out mice (chapter 4).

Page 225: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

224

This so-called cholinergic anti-inflammatory pathway is suggested to represent an

additional system controlling the inflammatory response to a wide range of threats to the

organism30. Inflammation is sensed by afferent nerve fibers and is subsequently relayed to

the brain. After integration of afferent information, the motor neurons of the vagus nerve are

activated and an integrated anti-inflammatory signal is sent back to the inflamed area. Still,

the presence of such a feedback loop (i.e. reflex) and its anatomical connections clearly

need to be demonstrated, and is currently being investigated. Nevertheless, this system

may represent an interesting new tool to contain undesired inflammatory processes. In

contrast to anti-inflammatory cytokines and the hormonal control by corticosteroids (HPA

axis), this neural system provides an integrated response that is lightning fast and localized.

Obviously, it may provide new therapeutic targets to control or dampen inflammation, not

only in case of sepsis or ileus, but most likely also in other inflammatory disorders like

rheumatoid arthritis and inflammatory bowel diseases.

Therapeutically these finding might have great impact. As we repeatedly have demonstrated

the importance of the local inflammatory response in the pathogenesis of postoperative

ileus, any therapeutic intervention preventing its occurrence could be an interesting

approach to treat this disorder. In the first chapters, we showed that interference with

adhesion molecules, necessary for leukocytes to leave the circulation and enter the area

of manipulation, either with antibodies or antisense oligonucleotides, indeed shortened the

ileus. Alternatively, we showed that interference with the release of mast cell mediators,

one of the first events in the pathophysiological cascade, is effective in our mouse model

(chapter 7). Ketotifen and doxantrazole, agents known to stabilize mast cells, prevented

handling induced inflammation and indeed shortened postoperative ileus in mice (chaper 7). Based on these findings, we designed a pilot proof-of-principle clinical study evaluating

the effect of ketotifen versus placebo treatment on postoperative gastric emptying in a

series of gynecological patients (chapter 9). Interestingly, we demonstrated that similar to

our animal experiments, ketotifen reduced the delay in gastric emptying evoked by surgery.

Although a larger study with a different dosing scheme is certainly required, this study

confirms our hypothesis in man and suggests that more specific mast cell stabilizers may

represent an interesting new approach to shorten postoperative ileus. Finally, interventions

that activate the cholinergic anti-inflammatory pathway might embody an attractive therapy.

Vagus nerve stimulation can be obtained either by electrical stimulation or administration

Page 226: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

10

Sum

mer

y an

d co

nclu

sions

225

of central application of drugs like CNI 1493, as shown in chapters 4 and 6. A much more

interesting approach would be to activate the vagus nerve by more physiological stimuli,

such as for example feeding. Recently, an interesting study was reported showing that a

meal containing high concentration of long-chain fatty acids activates vagal afferents via

endogenous cholecystokinin release31. In a model of hemorrhagic shock, feeding reduced

the production of TNF and the degree of inflammation and prevented the increase in

mucosal permeability. Based in these observations, we will study the potential beneficial

effect of early feeding of a high fat meal in the peri-operative period as potential treatment

of postoperative ileus. Finally, we showed that acetylcholine released by the vagus nerve

dampens the cytokine production of macrophages via binding to the alpha7 nicotinic

receptor (chapter 5). Drugs interacting with this receptor will mimic the effect of vagus

nerve stimulation and are theoretically interesting agents with potential anti-inflammatory

properties. Treatment with AR-R17779, a specific agonist to the alpha7 nicotinic receptor,

indeed prevented inflammation and shortened postoperative ileus in our mouse model

(chapter 5). Surprisingly though, the production of cytokines by macrophages in vitro was

only slightly reduced, in contrast to nicotine itself, indicating that other nicotinic receptors

and/or other cells may be involved explaining the in vivo effect. Nevertheless, clinical

studies evaluating the efficacy of alpha7 nicotinic receptor agonists are certainly warranted

and will be studied in the near future.

In summary, the data presented in the current thesis have provided substantial new insight

into the pathogenesis of prolonged postoperative ileus and have identified new therapeutic

targets. Our work is indirectly also a plea for minimal invasive surgery as our data clearly

indicate that intestinal handling during surgery should be avoided as much as possible.

Page 227: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

226

Reference ListPrasad M, Matthews JB. Deflating postoperative ileus. Gastroenterology 1999;117:489-492.1. Longo WE, Virgo KS, Johnson FE, Oprian CA, Vernava AM, Wade TP, Phelan MA, Henderson 2. WG, Daley J, Khuri SF. Risk factors for morbidity and mortality after colectomy for colon can-cer. Dis.Colon Rectum 2000;43:83-91.Collins TC, Daley J, Henderson WH, Khuri SF. Risk factors for prolonged length of stay after 3. major elective surgery. Ann.Surg. 1999;230:251-259.Boeckxstaens GE, Hirsch DP, Kodde A, Moojen TM, Blackshaw A, Tytgat GN, Blommaart PJ. 4. Activation of an adrenergic and vagally-mediated NANC pathway in surgery-induced fundic relaxation in the rat. Neurogastroenterol.Motil. 1999;11:467-474.De Winter BY, Boeckxstaens GE, De Man JG, Moreels TG, Herman AG, Pelckmans PA. 5. Effect of adrenergic and nitrergic blockade on experimental ileus in rats. Br.J.Pharmacol. 1997;120:464-468.De Winter BY, Boeckxstaens GE, De Man JG, Moreels TG, Schuurkes JA, Peeters TL, Herman 6. AG, Pelckmans PA. Effect of different prokinetic agents and a novel enterokinetic agent on postoperative ileus in rats. Gut 1999;45:713-718.Barquist E, Bonaz B, Martinez V, Rivier J, Zinner MJ, Tache Y. Neuronal pathways involved in 7. abdominal surgery-induced gastric ileus in rats. Am.J.Physiol 1996;270:R888-R894.Clevers GJ, Smout AJ. The natural course of postoperative ileus following abdominal surgery. 8. Neth J Surg 1989;41:97-9.Livingston EH, Passaro EP, Jr. Postoperative ileus. Dig.Dis.Sci. 1990;35:121-132.9. Holzer P, Lippe IT, Amann R. Participation of capsaicin-sensitive afferent neurons in gastric mo-10. tor inhibition caused by laparotomy and intraperitoneal acid. Neuroscience 1992;48:715-22.Plourde V, Wong HC, Walsh JH, Raybould HE, Tache Y. CGRP antagonists and capsaicin on 11. celiac ganglia partly prevent postoperative gastric ileus. Peptides 1993;14:1225-1229.Bonaz B, Plourde V, Tache Y. Abdominal surgery induces Fos immunoreactivity in the rat brain. 12. J.Comp Neurol. 1994;349:212-222.Schwenk W, Haase O, Neudecker J, Muller JM. Short term benefits for laparoscopic colorectal 13. resection. Cochrane.Database.Syst.Rev. 2005:CD003145.Chen HH, Wexner SD, Iroatulam AJ, Pikarsky AJ, Alabaz O, Nogueras JJ, Nessim A, Weiss 14. EG. Laparoscopic colectomy compares favorably with colectomy by laparotomy for reduction of postoperative ileus. Dis.Colon Rectum 2000;43:61-65.Veldkamp R, Kuhry E, Hop WC, Jeekel J, Kazemier G, Bonjer HJ, Haglind E, Pahlman L, 15. Cuesta MA, Msika S, Morino M, Lacy AM. Laparoscopic surgery versus open surgery for colon cancer: short-term outcomes of a randomised trial. Lancet Oncol. 2005;6:477-484.Kalff JC, Buchholz BM, Eskandari MK, Hierholzer C, Schraut WH, Simmons RL, Bauer AJ. Bi-16. phasic response to gut manipulation and temporal correlation of cellular infiltrates and muscle dysfunction in rat. Surgery 1999;126:498-509.Kalff JC, Carlos TM, Schraut WH, Billiar TR, Simmons RL, Bauer AJ. Surgically induced leuko-17. cytic infiltrates within the rat intestinal muscularis mediate postoperative ileus. Gastroenterol-ogy 1999;117:378-387.Foreman JC. Substance P and calcitonin gene-related peptide: effects on mast cells and in hu-18. man skin. Int Arch Allergy Appl Immunol 1987;82:366-71.Sharkey KA. Substance P and calcitonin gene-related peptide (CGRP) in gastrointestinal 19. inflammation. Ann N Y Acad Sci 1992;664:425-42.Suzuki R, Furuno T, McKay DM, Wolvers D, Teshima R, Nakanishi M, Bienenstock J. Direct 20. neurite-mast cell communication in vitro occurs via the neuropeptide substance P. J.Immunol. 1999;163:2410-2415.Berin MC, Kiliaan AJ, Yang PC, Groot JA, Kitamura Y, Perdue MH. The influence of mast cells 21. on pathways of transepithelial antigen transport in rat intestine. J.Immunol. 1998;161:2561-2566.

Page 228: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

10

Sum

mer

y an

d co

nclu

sions

227

Schwarz NT, Beer-Stolz D, Simmons RL, Bauer AJ. Pathogenesis of paralytic ileus: intestinal 22. manipulation opens a transient pathway between the intestinal lumen and the leukocytic infil-trate of the jejunal muscularis. Ann.Surg. 2002;235:31-40.Mikkelsen HB, Mirsky R, Jessen KR, Thuneberg L. Macrophage-like cells in muscularis externa 23. of mouse small intestine: immunohistochemical localization of F4/80, M1/70, and Ia-antigen. Cell Tissue Res. 1988;252:301-306.Kalff JC, Schraut WH, Simmons RL, Bauer AJ. Surgical manipulation of the gut elicits an intes-24. tinal muscularis inflammatory response resulting in postsurgical ileus. Ann.Surg. 1998;228:652-663.Kalff JC, Turler A, Schwarz NT, Schraut WH, Lee KK, Tweardy DJ, Billiar TR, Simmons RL, 25. Bauer AJ. Intra-abdominal activation of a local inflammatory response within the human mus-cularis externa during laparotomy. Ann.Surg. 2003;237:301-315.Borovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI, Watkins LR, Wang H, Abumrad N, 26. Eaton JW, Tracey KJ. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 2000;405:458-462.Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S, Li JH, Wang H, Yang H, Ulloa L, 27. Al Abed Y, Czura CJ, Tracey KJ. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature 2003;421:384-388.Borovikova LV, Ivanova S, Nardi D, Zhang M, Yang H, Ombrellino M, Tracey KJ. Role of vagus 28. nerve signaling in CNI-1493-mediated suppression of acute inflammation. Auton.Neurosci. 2000;85:141-147.Bernik TR, Friedman SG, Ochani M, DiRaimo R, Ulloa L, Yang H, Sudan S, Czura CJ, Ivanova 29. SM, Tracey KJ. Pharmacological stimulation of the cholinergic antiinflammatory pathway. J.Exp.Med. 2002;195:781-788.Tracey KJ. The inflammatory reflex. Nature 2002;420:853-859.30. Luyer MD, Greve JW, Hadfoune M, Jacobs JA, Dejong CH, Buurman WA. Nutritional stimu-31. lation of cholecystokinin receptors inhibits inflammation via the vagus nerve. J.Exp.Med. 2005;202:1023-1029.

Page 229: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

11

Page 230: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

11C

hapt

er 1

1Samevatting en Conclusies

Dankwoord

Colour Figures

Page 231: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

230

MSamenvatting en conclusiesMeer dan een eeuw na de eerste beschrijvingen in de wetenschappelijke literatuur is

postoperatieve ileus nog steeds een frequent voorkomend medisch probleem met niet

onaanzienlijke morbiditeit en sociaal-economische consequenties1, 2. Tot op de dag van

vandaag blijft iedere patiënt die een buikoperatie ondergaat tot enkele dagen na de ingreep

in het ziekenhuis opgenomen in verband met klachten van misselijkheid, gebrek aan

eetlust, niet kunnen eten en het uitblijven van ontlasting3. Zowel dierexperimenteel-4-7 als

patiëntgebonden-onderzoek1, 8 hebben aangetoond dat deze symptomen, beter bekend als

postoperatieve ileus, het gevolg zijn van afwezige of gestoorde maag-, darm motoriek en

het onvermogen van de darmen om hun inhoud voort te stuwen. Voornamelijk pijnprikkels

gedurende de operatieve ingreep, opgewekt door de huid incisie, het openen van buikholte

maar voornamelijk door het aanraken van de darmen gedurende de procedure, blijken

belangrijke veroorzakers te zijn van deze instantane paralyse van het spijsverteringskanaal4,

9, 10. Tot voor kort werd de activatie van inhiberende zenuwbanen als gevolg van activatie

van pijn- en mechano-sensoren, beschouwd als belangrijkste onderliggende oorzaak.

Farmacologische neuronale blokkade, klieving van spinale zenuwbanen, depletie van

afferente zenuwbanen met capsaicine en identificatie van de betrokken zenuwbanen

en hersenencentra, bevestigen allen deze hypothese4, 5, 7, 11, 12. De meeste van deze

experimenten zijn echter tijdens de operatie uitgevoerd of hebben alleen het effect op de

maag- darm motoriek bestudeerd gedurende de 1e 3uur na de ingreep. Inmiddels weten

we echter dat deze “vroege fase” slechts een fractie van het klinische probleem is, vooral

omdat postoperatieve ileus veel langer aanhoudt en enkele dagen duurt. In dit proefschrift

beschrijven we inderdaad dat postoperative ileus vooral bepaald wordt door een latere en

langdurige fase die veroorzaakt wordt ontsteking van de darm.

Gedurende operatieve ingrepen in de buikholte is het hanteren van darmlissen onvermijdelijk,

of het nu is om het doelorgaan te bereiken of om de darm te inspecteren voor afwijkingen.

Alhoewel de exacte initiële prikkel onopgehelderd blijft, is het inmiddels duidelijk geworden

dat de mate van darmmanipulatie gedurende een operatie een belangrijke voorspeller kan

zijn voor de ernst van de postoperatieve ileus. Deze observatie heeft dan ook ongetwijfeld

bijgedragen aan de ontwikkeling van minimaal invasieve chirurgische technieken. Onderzoek

Page 232: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

11

Sam

enva

tting

en

conc

lusie

s,

dank

woo

rd e

n co

lour

figu

res

231

heeft aangetoond dat deze relatief nieuwe wijze van opereren inderdaad resulteert in

verkorting van de duur van postoperatieve ileus en ziekenhuis opname13-15. Het is echter

pas sinds enkele jaren dat we zijn gaan beginnen te begrijpen hoe darmmanipulatie kan

leiden tot aanhoudende neuronmusculaire dysfunctie. Kalff en collega’s hebben aangetoond

dat 3 tot 4 uur na het hanteren van dunne darm lissen (darmmanipulatie) de spierlaag in

de darmwand ontstoken raakt16. In knaagdieren leidt deze locale ontsteking tot vertraging

van de darmtransit (voortstuwen van darminhoud) en verminderde spierfunctie17. Deze

bevindingen hebben geresulteerd in de ontstekingshypothese die stelt dat de persisterende

fase van postoperatieve ileus veeleer het gevolg is van manipulatie geïnduceerde ontsteking

en als gevolg hiervan gestoorde maag-, darmmotoriek door activatie van inhibitoire

zenuwbanen. Op basis van deze observaties kunnen we de pathofysiologie nu indelen in

2 fasen; een acute, kortdurende, neurogene fase en een 2e late inflammatoire fase (vanaf

3 uur na chirurgie). Gezien de duur is deze 2e fase vanuit klinisch oogpunt veruit de meest

belangrijke van de twee. In dit proefschrift hebben we ons dan ook gericht op het verder

ontrafelen van het onderliggende cellulaire mechanisme met als doel de (preventieve)

behandeling van postoperatieve ileus te verbeteren.

Hoewel de inflammatoire theorie verklaart hoe de maag-, en darmmotoriek gestoord blijft

na beëindiging van de chirurgische ingreep, blijft het onduidelijk waarom de peristaltiek van

gans de gastrointestinale tractus verstoord is. Aannemende dat darmmanipulatie resulteert

in een locale ontsteking van de darmspierlaag blijft het onduidelijk hoe dit leidt tot gestoorde

propulsieve functie van die delen van het maagdarmstelsel die niet gemanipuleerd zijn.

Eén verklaring zou kunnen zijn dat manipulatie gedurende de chirurgie resulteert in een

gegeneraliseerde ontsteking. Echter experimenten in ons laboratorium hebben uitgewezen

dat 24 uur na abdominale chirurgie alleen die segmenten ontstoken zijn die gemanipuleerd

zijn geweest gedurende de procedure. Om het mechanisme verder te onderzoeken dat

leidt tot gegeneraliseerde verstoring van maag-, en darmmotoriek hebben we daarom een

muismodel ontwikkeld waarin 24uur na abdominale chirurgie de maagontledigingssnelheid

bepaald wordt als maat van gegeneraliseerde ileus (hoofdstuk 2). In deze experimenten

wordt in de ene groep de dunne darm gedurende 5 minuten voorzichtig gemanipuleerd

terwijl in de controle groep alleen de buikholte wordt geopend (laparotomie). Vervolgens

worden de darmlissen weer voorzichtig in de buikholte teruggeplaatst waarna de buikholte

wordt gesloten en 24 uur later wordt de maagontledigingssnelheid bepaald. De dieren die

Page 233: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

232

darmmanipulatie ondergingen tijdens de operatie toonden een significante vertraging van de

maagontlediging ten opzichte van controle dieren (hoofdstuk 2). Ook de ontstekingsreactie

in spierlaag van de darmwand was alleen aantoonbaar in muizen die darmmanipulatie

hadden ondergaan (hoofdstuk 2). Belangrijk hierbij is te vermelden dat deze ontsteking

alleen aanwezig was in die segmenten die waren gemanipuleerd tijdens de operatie.

Desalniettemin was tot 48 uur na de operatie de maaglediging significant vertraagd in

deze dieren (hoofdstuk 2). Daarnaast hebben we ook ontdekt dat het voorkomen van

de manipulatie gemedieerde ontsteking door middel behandeling met oa. antilichamen of

antisense oligonucleotiden gericht tegen het adhesie molecuul ICAM-1 (belangrijk bij de

rekrutering van ontstekingscellen vanuit de bloedsomloop) resulteert in de normalisatie

van maagontlediging (hoofdstuk 3).

Hoe kan locale ontsteking van de dunne darm leiden tot vertraging van de maagontlediging?

We veronderstelden dat neuronale reflexbanen geactiveerd raken door het locale

ontstekingsinfiltraat. Om deze hypothese te onderzoeken werden dieren voorbehandeld

met de ganglionerge blokkers guanethidine en hexamethonium(hoofdstuk 2). De

resultaten van dit experiment toonde inderdaad een normalisering van de maagfunctie

terwijl de locale ontsteking in de dunne darm nog wel aanwezig was. Om de aanwezigheid

van een inhiberende zenuwreflex in de pathofysiologie van postoperatieve ileus verder

te onderzoeken hebben we vervolgens het ruggemerg gekleurd voor c-fos (een zenuw

activatie marker) (hoofdstuk 2). Dieren die darmmanipulatie hadden ondergaan maar

niet de controle muizen toonden c-fos expressie in het ruggenmerg, wat onze hypothese

andermaal bevestigde (hoofdstuk 2). Op basis van deze resultaten concluderen we

dat de aanhoudende (late) fase in postoperatieve ileus het gevolg is van neuro-immuun

interactie tussen de ontstekingscellen in de gemanipuleerde darmsegment en de afferente

(sensorische) innnervatie van het maag-, en darmstelsel. Dit samenspel activeert

vervolgens adrenerge zenuwbanen die via het ruggemerg het gehele maag-, darmstelsel

negatief beïnvloeden.

Het volgende vraagstuk was op welke wijze darmmanipulatie leidt tot een locale

ontstekingsreactie met rekrutering van ontstekingscellen. Natuurlijk kan weefselschade

die het gevolg is van manipulatie zorgen voor de attractie van ontstekingscellen en dus

bijdragen tot de totstandkoming van een locale ontstekingsreactie. Onze gedachte was

Page 234: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

11

Sam

enva

tting

en

conc

lusie

s,

dank

woo

rd e

n co

lour

figu

res

233

echter dat sensorische zenuwvezels een belangrijkere rol zouden kunnen spelen, mede

gezien het feit dat eerdere studies hebben laten zien dat intensieve stimulatie van afferente

zenuwvezels een belangrijke prikkel vormen voor de ontwikkeling van locale inflammatie,

ook wel neurogene inflammatie genoemd18, 19. Afferente zenuwvezels stellen neuropeptiden

zoals Calcitonine Gene Related Peptide (CGRP) en substance P vrij wanneer ze intens

worden geactiveerd19. Deze eiwitten zijn potente pro-inflammatoire mediatoren, vooral

door hun vermogen om mestcellen te activeren18, 20. Het vrijstellen van mediatoren door

mestcellen heeft niet alleen een direct pro-inflammatoir effect maar resulteert ook in een

kortstondig verhoogde permeabiliteit (doorlaatbaarheid) van het darmslijmvlies (mucosa)21.

Dit laatste maakt het voor bacteriën mogelijk de darmwand te penetreren, een fenomeen

dat al eerder beschreven is ten gevolge van darmmanipulatie22. Schwarz et al. hebben

laten zien dat darmmanipulatie leidt tot de influx van luminale micropartikels naar de

darmwand ongeveer 4 uur na manipulatie22. Zoals gezegd kunnen bacteriën ten gevolge

van deze tijdelijke opening van de mucosale barrière, vanuit het darmlumen de darmwand

penetreren en vormen daar een belangrijke stimulus voor het immuunsysteem. Macrofagen

die als een soort netwerk van poortwachters tussen de longitudinale en circulaire spierlaag

van de darm liggen23, worden enkele uren na darmmanipulatie geactiveerd zoals onder

andere blijkt uit de toename van IL-6, iNOS en LFA-1concentraties17, 24, 25.

In hoofdstuk 7 hebben we de belangrijke rol die mestcellen vervullen in de pathofysiologie

van postoperatieve ileus aangetoond door aan te tonen dat manipulatie geïnduceerde

ontsteking en ileus gereduceerd zijn in muizen die voorbehandeld zijn met mestcel

stabilisatoren of die mestcel deficiënt zijn (W/Wv muizen). Mestcel reconstitutie in deze

W/Wv muizen herstelt de ontstekingsrespons. Ook in patiënten hebben wij het vrijkomen

van mestcel mediatoren in de buikholte aansluitend op subtiele darmmanipulatie kunnen

aantonen (hoofdstuk 8). Vergelijkbaar met onze observaties in muizen resulteert mestcel

activatie ook bij de mens in het vrijkomen of opreguleren van ontstekingsmediatoren zoals

IL-6, IL-8, iNOS en ICAM-1. Dit proces leidt uiteindelijk ook hier tot de rekrutering van

ontstekingscellen naar de spierlaag van de darm. Opvallend hierbij is overigens dat dit

proces nagenoeg alleen waarneembaar is in patiënten die een conventionele open buik

operatie (laparotomie) ondergaan en niet in patiënten die een minimaal invasieve ingreep

ondergingen. Ook visualisatie van ontstekingscelrekrutering 24 uur voor en na chirurgie,

middels het markeren van witte bloedcellen met een radioactieve merkstof (leukocyten

Page 235: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

234

SPECT scintigrafie), toonde vergelijkbare resultaten (hoofdstuk 8). Deze resultaten tonen

duidelijk aan dat zowel in proefdieren als in mensen, mestcel activatie ten gevolge van

chirurgische darmmanipulatie een belangrijke eerste stap vormt in de cascade die leidt tot

locale darmontsteking en postoperatieve ileus.

Zoals weergegeven in Summarizing figure (zie kleuren katern) gaat mestcel gemedieerde

toename van darm permeabiliteit gepaard met microbiële translocatie. Dit laatste fenomeen

is waarschijnlijk verantwoordelijk voor de eerder door Kalff et al. beschreven activatie

van het netwerk van macrofagen gelegen tussen de darmspierlagen24. Interessant

hierbij is dat Borovikova et al. hebben aangetoond dat stimulatie van de nervus vagus

de activatie van macrofagen kan doen verminderen26. In een experimenteel sepsis model

hebben deze onderzoekers aangetoond dat elektrische stimulatie van de nervus vagus

een betere overleving en bloeddruk controle tonen na infusie van LPS. Acetylcholine, de

neurotransmitter vrijgesteld door de nervus vagus, bindt aan alfa7 nicotinerge receptor

op macrofagen27 met verminderde vrijstelling van pro-inflammatoire mediatoren zoals

TNF-alfa27. Aangezien het maag-, darmstelsel overwegend onder de controle staat van

de nervus vagus, hebben wij onderzocht of elektrische stimulatie van de nervus vagus

ook de activatie van macrofagen in de darmwand kan beïnvloeden om op deze wijze de

ontstekingreactie en ileus na darmmanipulatie te verminderen (hoofdstuk 4). Hieruit is

gebleken dat elektrische stimulatie van de vagus intra-peritoneale vrijstelling van TNF,

MIP-2 en IL-6 3 uur postoperatief inderdaad kan verminderen in ons model, een maat

voor verminderde macrofaag activatie. Bovendien worden er minder ontstekingscellen

gerekruteerd wat resulteerde in een normalisatie van de maagontlediging (hoofdstuk4). Om

de rol van dit anti-inflammatoire mechanisme verder te exploreren hebben we vervolgens

experimenten uitgevoerd met CNI-1493, een MAPKinase remmer die vagus afhankelijke

anti-inflammatoire eigenschappen heeft28, 29. Net als elektrische stimulatie van de nervus

vagus vermindert intraventriculaire toediening van CNI-1493 de ontsteking en verbetert het

de maagontledigingsfunctie, een effect dat te niet wordt gedaan door vagotomie (hoofdstuk 6). Deze serie van proeven heeft aangetoond dat de nervus vagus ook in het maag-,

darmstelsel een belangrijke regulatoire invloed heeft op het immuunsysteem. Vervolgens

hebben we in geïsoleerde macrofagen aangetoond dat acetycholine macrofaagactivatie

remt via de alfa7 nicotine receptor (hoofdstuk 4) en het Jak2/STAT3 signaleringspad. Het

belang van dit signaleringsmechanisme wordt benadrukt door het feit dat vagus stimulatie

Page 236: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

11

Sam

enva

tting

en

conc

lusie

s,

dank

woo

rd e

n co

lour

figu

res

235

de ontstekingsrespons niet kan onderdrukken in STAT3 geconditioneerde knock-out

muizen (Hoofdstuk 4).

Dit zogenaamde cholinerge anti-inflammatoire pad wordt beschouwd als een additioneel

regulatoir systeem van het immuunsysteem30. Hierin wordt de ontsteking gedetecteerd door

sensibele zenuwbanen en doorgegeven aan het brein. Na de verwerking van deze afferente

informatie worden de motorneuronen van de vagus geactiveerd en wordt er een geïntegreerd

anti-inflammatoir signaal teruggestuurd naar het ontstoken gebied. Echter het bestaan van

een dergelijk controle systeem (reflex) en betrokken anatomische verbindingen moeten

nog daadwerkelijk worden aangetoond. In tegenstelling tot anti-inflammatoire cytokinen en

hormonale regulatie middels corticosteroïden (via de HPA-as) zorgt dit neuronale syteem

voor een geïntegreerde respons die extreem snel en locatie specifiek is. Dit concept zal

ongetwijfeld resulteren in de ontwikkeling van nieuwe behandelstrategieën die niet alleen

toepasbaar zijn in sepsis of ileus maar ook in een scala aan andere ontsteking gerelateerde

aandoeningen.

De hier gepresenteerde resultaten hebben mogelijk belangrijke therapeutische gevolgen.

Gezien het feit dat we bij herhaling hebben laten zien dat de locale inflammatoire respons

belangrijk is in de pathogenese van postoperatieve ileus is iedere interventie die deze

respons kan voorkomen in beginsel een interessante therapeutische optie. In de eerste

hoofdstukken hebben we laten zien dat interventie op het niveau adhesie moleculen, nodig

bij de rekrutering van ontstekingscellen vanuit de bloedsomloop naar het ontstoken weefsel,

doormiddel van antilichamen of antisense oligonucleotiden het beloop van postoperatieve

ileus gunstig kunnen beïnvloeden. Daarnaast hebben we ook laten zien dat het voorkomen

van mestcel degranulatie (vrijkomen van mestcel specifieke pro-inflammatoire eiwitten), een

van de eerste processen in de pathofysiologische cascade, een gunstig effect heeft op het

beloop van postoperatieve ileus (hoofdstuk 7). Ketotifen en doxantrazole, twee farmaca

die bekend staan als mestcel stabiliserende agens, voorkomen manipulatie gemedieerde

ontsteking en verkorten het beloop van postoperatieve ileus in muizen (hoofdstuk 7). Op

basis van deze resultaten hebben we een pilot-studie ontworpen waarin we het concept

van mestcel stabilisatie als behandeling voor postoperatieve ileus hebben onderzocht. In

deze studie hebben we op een dubbelblind gerandomiseerde wijze gekeken naar het effect

van ketotifen behandeling ten opzichte van placebo op de postoperatieve maagontlediging

Page 237: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

236

in een gynaecologische patiëntenpopulatie (hoofdstuk 9). Naar analogie met ons

dierexperimenteel werk was de maagontlediging sneller na behandeling met ketotifen.

Alhoewel een grotere studie mogelijk met een ander doseringsschema nodig is, bevestigt

deze studie onze hypothese en suggereert dat meer specifieke mestcel stabilisatoren een

attractieve behandel optie zouden kunnen vormen om postoperatieve ileus te voorkomen.

Tot slot kunnen farmaca die het cholinerge anti-inflammatoire mechanisme activeren een

interessante benadering zijn om postoperatieve ileus te verkorten. Dit zou kunnen worden

bewerkstelligd door middel van elektrische stimulatie van de nervus vagus of toediening

van farmaca zoals CNI-1493 (beschreven in hoofdstuk 4 en 6). Een veel elegantere en

meer fysiologische methode van vagus activatie is voeding. Een interessante recente

publicatie heeft aangetoond dat voeding die hoge concentratie lange-keten vetzuren

bevat afferente vagale zenuwvezels activeert door middel van endogene cholecystokinine

vrijstelling31. In een model voor hemorhagische shock hebben deze auteurs aangetoond dat

voeding de productie van TNF vermindert, de ontstekingsreactie dempt en toename van de

darmpermeabiliteit voorkomt. Gebaseerd op deze gegevens willen wij het effect van vroege

voeding met vetrijke maaltijden in de peri-operatieve fase op het beloop van postoperatieve

ileus gaan bestuderen. In hoofdstuk 5 hebben we al laten zien dat acetylcholine, vrijgesteld

door de nervus vagus, de cytokine vrijstelling door macrofagen vermindert via alfa7 nicotine

receptor binding. Agonisten voor deze receptor bootsen het effect van vagusstimulatie na

en zijn in theorie potente anti-inflammatoire medicijnen. Behandeling met AR-R17779, een

specifieke alfa7 nicotine receptor agonist, vermindert inderdaad de inflammatoire respons

en verbetert de maagledigingsfunctie in ons postoperatieve ileus model (hoofdstuk 5).

Vreemd genoeg is het cytokine productie reducerende vermogen van dit middel in stimulatie

proeven slechts minimaal. Dit in tegenstelling tot het effect van nicotine wat suggereert dat

ander nicotine receptoren en/of celtypen betrokken zijn in dit proces. Desalniettmin zijn

klinische studies naar het effect van alfa7 nicotinerge agonisten gerechtvaardigd en zullen

zeker in de nabije toekomst worden uitgevoerd.

Page 238: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

11

Sam

enva

tting

en

conc

lusie

s,

dank

woo

rd e

n co

lour

figu

res

237

Samenvattend kunnen we stellen dat de gegevens gepresenteerd in dit proefschrift een

schat aan nieuwe inzichten heeft gegenereerd met betrekking tot de pathofysiologie van

postoperatieve ileus en hierbij meerdere nieuwe therapeutische targets heeft geidentificeerd.

Vooral ook omdat we duidelijk hebben aangetoond dat darmmanipulatie gedurende

heelkundige ingrepen zoveel mogelijk dient te worden vermeden, is dit proefschrift tevens

een indirect pleidooi voor de verdere ontwikkeling van minimaal invasieve chirurgische

technieken.

Page 239: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

238

Referentie Lijst Prasad M, Matthews JB. Deflating postoperative ileus. Gastroenterology 1999;117:489-492.1. Longo WE, Virgo KS, Johnson FE, Oprian CA, Vernava AM, Wade TP, Phelan MA, Henderson 2. WG, Daley J, Khuri SF. Risk factors for morbidity and mortality after colectomy for colon can-Risk factors for morbidity and mortality after colectomy for colon can-cer. Dis.Colon Rectum 2000;43:83-91.Collins TC, Daley J, Henderson WH, Khuri SF. Risk factors for prolonged length of stay after 3. major elective surgery. Ann.Surg. 1999;230:251-259.Boeckxstaens GE, Hirsch DP, Kodde A, Moojen TM, Blackshaw A, Tytgat GN, Blommaart PJ. 4. Activation of an adrenergic and vagally-mediated NANC pathway in surgery-induced fundic relaxation in the rat. Neurogastroenterol.Motil. 1999;11:467-474.De Winter BY, Boeckxstaens GE, De Man JG, Moreels TG, Herman AG, Pelckmans PA. 5. Effect of adrenergic and nitrergic blockade on experimental ileus in rats. Br.J.Pharmacol. 1997;120:464-468.De Winter BY, Boeckxstaens GE, De Man JG, Moreels TG, Schuurkes JA, Peeters TL, Herman 6. AG, Pelckmans PA. Effect of different prokinetic agents and a novel enterokinetic agent on postoperative ileus in rats. Gut 1999;45:713-718.Barquist E, Bonaz B, Martinez V, Rivier J, Zinner MJ, Tache Y. Neuronal pathways involved in 7. abdominal surgery-induced gastric ileus in rats. Am.J.Physiol 1996;270:R888-R894.Clevers GJ, Smout AJ. The natural course of postoperative ileus following abdominal surgery. 8. Neth J Surg 1989;41:97-9.Livingston EH, Passaro EP, Jr. Postoperative ileus. Dig.Dis.Sci. 1990;35:121-132.9. Holzer P, Lippe IT, Amann R. Participation of capsaicin-sensitive afferent neurons in gastric mo-10. tor inhibition caused by laparotomy and intraperitoneal acid. Neuroscience 1992;48:715-22.Plourde V, Wong HC, Walsh JH, Raybould HE, Tache Y. CGRP antagonists and capsaicin on 11. celiac ganglia partly prevent postoperative gastric ileus. Peptides 1993;14:1225-1229.Bonaz B, Plourde V, Tache Y. Abdominal surgery induces Fos immunoreactivity in the rat brain. 12. J.Comp Neurol. 1994;349:212-222.Schwenk W, Haase O, Neudecker J, Muller JM. Short term benefits for laparoscopic colorectal 13. resection. Cochrane.Database.Syst.Rev. 2005:CD003145.Chen HH, Wexner SD, Iroatulam AJ, Pikarsky AJ, Alabaz O, Nogueras JJ, Nessim A, Weiss 14. EG. Laparoscopic colectomy compares favorably with colectomy by laparotomy for reduction of postoperative ileus. Dis.Colon Rectum 2000;43:61-65.Veldkamp R, Kuhry E, Hop WC, Jeekel J, Kazemier G, Bonjer HJ, Haglind E, Pahlman L, 15. Cuesta MA, Msika S, Morino M, Lacy AM. Laparoscopic surgery versus open surgery for colon cancer: short-term outcomes of a randomised trial. Lancet Oncol. 2005;6:477-484.Kalff JC, Buchholz BM, Eskandari MK, Hierholzer C, Schraut WH, Simmons RL, Bauer AJ. Bi-16. phasic response to gut manipulation and temporal correlation of cellular infiltrates and muscle dysfunction in rat. Surgery 1999;126:498-509.Kalff JC, Carlos TM, Schraut WH, Billiar TR, Simmons RL, Bauer AJ. Surgically induced leuko-17. cytic infiltrates within the rat intestinal muscularis mediate postoperative ileus. Gastroenterol-ogy 1999;117:378-387.Foreman JC. Substance P and calcitonin gene-related peptide: effects on mast cells and in hu-18. man skin. Int Arch Allergy Appl Immunol 1987;82:366-71.Sharkey KA. Substance P and calcitonin gene-related peptide (CGRP) in gastrointestinal 19. inflammation. Ann N Y Acad Sci 1992;664:425-42.Suzuki R, Furuno T, McKay DM, Wolvers D, Teshima R, Nakanishi M, Bienenstock J. Direct 20. neurite-mast cell communication in vitro occurs via the neuropeptide substance P. J.Immunol. 1999;163:2410-2415.Berin MC, Kiliaan AJ, Yang PC, Groot JA, Kitamura Y, Perdue MH. The influence of mast cells 21. on pathways of transepithelial antigen transport in rat intestine. J.Immunol. 1998;161:2561-2566.

Page 240: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

11

Sam

enva

tting

en

conc

lusie

s,

dank

woo

rd e

n co

lour

figu

res

239

Schwarz NT, Beer-Stolz D, Simmons RL, Bauer AJ. Pathogenesis of paralytic ileus: intestinal 22. manipulation opens a transient pathway between the intestinal lumen and the leukocytic infil-trate of the jejunal muscularis. Ann.Surg. 2002;235:31-40.Mikkelsen HB, Mirsky R, Jessen KR, Thuneberg L. Macrophage-like cells in muscularis externa 23. of mouse small intestine: immunohistochemical localization of F4/80, M1/70, and Ia-antigen. Cell Tissue Res. 1988;252:301-306.Kalff JC, Schraut WH, Simmons RL, Bauer AJ. Surgical manipulation of the gut elicits an intes-24. tinal muscularis inflammatory response resulting in postsurgical ileus. Ann.Surg. 1998;228:652-663.Kalff JC, Turler A, Schwarz NT, Schraut WH, Lee KK, Tweardy DJ, Billiar TR, Simmons RL, 25. Bauer AJ. Intra-abdominal activation of a local inflammatory response within the human mus-cularis externa during laparotomy. Ann.Surg. 2003;237:301-315.Borovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI, Watkins LR, Wang H, Abumrad N, 26. Eaton JW, Tracey KJ. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 2000;405:458-462.Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S, Li JH, Wang H, Yang H, Ulloa L, 27. Al Abed Y, Czura CJ, Tracey KJ. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature 2003;421:384-388.Borovikova LV, Ivanova S, Nardi D, Zhang M, Yang H, Ombrellino M, Tracey KJ. Role of vagus 28. nerve signaling in CNI-1493-mediated suppression of acute inflammation. Auton.Neurosci. 2000;85:141-147.Bernik TR, Friedman SG, Ochani M, DiRaimo R, Ulloa L, Yang H, Sudan S, Czura CJ, Ivanova 29. SM, Tracey KJ. Pharmacological stimulation of the cholinergic antiinflammatory pathway. J.Exp.Med. 2002;195:781-788.Tracey KJ. The inflammatory reflex. Nature 2002;420:853-859.30. Luyer MD, Greve JW, Hadfoune M, Jacobs JA, Dejong CH, Buurman WA. Nutritional stimu-31. lation of cholecystokinin receptors inhibits inflammation via the vagus nerve. J.Exp.Med. 2005;202:1023-1029.

Page 241: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

240

HDankwoordHet einde nadert. Een sentimenteel moment van overpeinzing maakt zich meester van

de auteur. Onvermijdelijk denkt hij op zo’n moment even terug aan wat hij nu eigelijk de

afgelopen jaren allemaal heeft uitgespookt.

Ik beschouw mijn onderzoeksperiode als één groot speelkwartier waarbij het AMC de

speeltuin was waar ik me als een kind in luilekkerland heb kunnen uitleven. Het is dan ook

met gemengde gevoelens dat ik een slotwoord op papier zet.

Dit wetenschappelijke avontuur zou ik nooit hebben kunnen volbrengen zonder de steun

van velen. Zij die al jaren deel uit maakten van mijn leven en mij hebben bijgestaan tijdens

de high’s en low’s die deze onderzoeksjaren met zich mee hebben gebracht. Velen van jullie

heb ik mogelijk tekort gedaan in mijn misschien wel bijna echocentrische preoccupatie met

dit boeiende maar soms ook zeer frustrerende werk. Ik ben jullie eeuwig dankbaar voor de

onvoorwaardelijke vriendschap die jullie mij hebben gegeven. Door het multidisciplinaire

karakter van het onderzoeksproject heb ik ook vele nieuwe mensen leren kennen. Ik

beschouw het als een groot voorrecht dat ik in de keuken van verscheidene disciplines en

instituten heb mogen snuffelen en ben dankbaar voor de gastvrijheid die men mij daarbij

geboden heeft. De inspiratie die het geeft om met mensen met verschillende expertise van

gedachte te wisselen over het onderzoek en meer..., heeft zeker bijgedragen aan het grote

plezier dat ik aan het doen van onderzoek heb beleeft.

Alhoewel ik het hier misschien wel het liefste bij zou willen laten, uit vrees in mijn

dankbetuiging te kort te schieten, ontkom ook ik er niet aan om een aantal mensen in

het bijzonder te noemen. Toch wil ik vanuit de grond van mijn hart hier alvast iedereen

bedanken die op welke wijze dan ook aan het tot stand komen van dit proefschrift heeft

bijgedragen!

Professor Dr. Boeckxstaens, beste Guy. Mijn wetenschappelijke mentor en spellingscontrole.

Jij hebt me wegwijs gemaakt in de wereld van neurogastroenterologie en wetenschap.

Ik beschouw het als een eer om onder jou te hebben mogen promoveren. Onder jouw

gedreven leiderschap heb ik een kleine eigenzinnige onderzoeksgroep zien uitgroeien

Page 242: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

11

Sam

enva

tting

en

conc

lusie

s,

dank

woo

rd e

n co

lour

figu

res

241

tot een autoriteit en heb getuigen mogen zijn van meerdere grootse wetenschappelijke

momenten waarvan velen alleen kunnen dromen. Ik dank je voor de mogelijkheden die je

me in de afgelopen jaren hebt geboden.

Dr. de Jonge, beste Wouter. Je bent op vele wijzen een onnavolgbaar voorbeeld voor me

geweest. Je ambitie en gedrevenheid zijn fenomenaal. Met veel bewondering heb ik vaak

gedwee aanschouwd hoe jij gedreven door je oprechte wetenschappelijke nieuwsgierigheid

met alles en iedereen een gesprek aanknoopte om iets te realiseren of om het naatje

van de kous, ten aanzien van een onderwerp, te weten te komen. Ondanks deze enorme

drive was er altijd tijd voor wat slap geouwehoer, een goed gesprek of flauwe “de Jonge”

grappen. Je enthousiasme is aanstekelijk en heeft me meer dan eens gemotiveerd. Waar

velen van je collega’s het lab verruilen voor een werkkamer ben jij niet uit het lab te slaan.

Tussendoor schrijf je daarnaast dan nog even de ene succesvolle subsidieaanvraag na

de andere, iets wat ik in je bewonder. Inmiddels heb je al je eigen onderzoeksgroep en is

je benoeming tot hoogleraar mijns inziens slechts een kwestie van tijd. Ik ben blij dat je

mijn co-promotor bent en hoop dat de toekomst ons weer samenbrengt om “belangrijke

enigmata” te ontrafelen.

Het motiliteitscentrum op C2, het kloppend hart. Lieve Aaltje, zonder jou was de ketotifen-

trial nooit wat geworden. Met veel plezier denk ik terug aan onze samenwerking en leuke

gesprekken. Je bent meer dan een fijne collega en ik hoop dat we snel weer eens tijd

kunnen vrijmaken om onder het genot van een hapje en drankje de wereldproblematiek

door te nemen. Sjoerd en Bram, toen C2-310 nog een mannenkamer was... Met jullie is

mijn promotie avontuur begonnen. Sjoerd, jammer genoeg ging jij al snel naar het Lucas

Andreas Ziekenhuis. Dank voor het bijbrengen van de fijne kneepjes van het ano-rectaal

functie onderzoek. Gelukkig zijn onze wegen na je vertrek al meer dan eens gekruist en

komen we elkaar zeker nog tegen. Bram, met jou heb ik lange tijd lief en leed gedeeld. Je

onuitputtelijke geduld en sociale instelling bewonder ik enorm. Dank voor je vriendschap

en de leuke tijd samen. Laten we snel het al lang geleden aan elkaar beloofde biertje

gaan drinken! Cynthia, helaas, maar niet onverwacht, heb je de motiliteit verruild voor een

nieuwe werkgever. Dank voor je ondersteuning en oplossend vermogen. Tamira, dank voor

de leuke gesprekken, het meedenken en het geven van je oprechte mening. Hanneke,

altijd in voor iets leuks. Dank voor de gezellige tijd samen, ik zal nooit vergeten hoe we

Page 243: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

242

samen op het Rembrandtplein lagen! Rene, amice! Rots in de branding, kamergenoot om

vijf voor twaalf en kritisch oor. De (ten onrechte) soms te stille (lees bescheiden) kracht van

de motiliteit. De leuke en inzichtelijke gesprekken met jou waren onbetaalbaar! Dennis,

dank voor je labsupport en soms bijna niet te volgen gevoel voor humor. Ik hoop dat je het

naar je zin hebt op je nieuwe werk. De vagus-girls, Esmerij en Susan. Jammer genoeg was

onze samenwerking relatief kort maar wel gezellig. Heel veel succes met jullie onderzoek,

dank! Ramona, Olaf (je hebt een moedige en goede beslissing genomen), Sjoerd B (the

next generation), Breg, Cathy en de poep-poli boys and girls (Mark, Fleur, Wieger, Maartje,

Michiel, Marloes, Noor en Olivia): allen dank voor de leuke tijd samen!

Dr. Bennink, beste Roel. Het was fantastisch om met je te mogen werken. Ik dank je voor

je onuitputtelijke vindingrijkheid, meedenkend vermogen en behulpzaamheid. Niets was

onmogelijk! Natuurlijk wil ik ook Formijn, Cynara, Jan, Ilse, Marsha en het hele team van de

Nucleaire Geneeskunde bedanken. Zonder jullie inzet (soms zelfs in het weekend!) waren

mijn experimenten en de klinische studies nooit het succes geworden wat het nu is!

Mijn steun en toeverlaat in het lab, Angelique! Bijna altijd goed gehumeurd en geïnteresseerd

in hoe het met je medemens gaat. We hebben elkaar leren kennen toen je, met veel

tegenzin, van G1 naar G2 moest verhuizen. Je hebt je ontpopt tot een top analist die

iedereen een helpende hand biedt. Inmiddels heb je een cardioloog aan de haak geslagen

en ben je moeder geworden van een lieve dochter: alle ingrediënten voor het geluk. Helaas

zien wij elkaar te weinig sinds ik het AMC verlaten heb. Dank voor je hulp en vriendschap.

Hoop snel weer eens bij Arko, Meike en jou te kunnen komen buurten.

Professor Buijs, beste Ruud, Jan en Caroline. Dank voor de goede en vooral ook gezellige

samenwerking op het NIH (tegenwoordig NIN). Jullie expertise was onmisbaar in de

“neuro-immuun interactie”. Inmiddels zijn jullie allen elders gaan werken. Ik wens jullie heel

veel succes en geluk in de nieuwe omgeving. Jan, het spijt me dat ik de minimale 1.5 x

Balkenende niet hebben kunnen realiseren.

Dr. te Velden, beste Anje. Onder jouw toeziend oog heb ik de eerste voorzichtige schreden

de wetenschap gezet. Zonder jou was ik waarschijnlijk nooit in contact gekomen met

Wouter et al., dank!

Page 244: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

11

Sam

enva

tting

en

conc

lusie

s,

dank

woo

rd e

n co

lour

figu

res

243

Dr. Buist, beste Marrije. Dank voor je inzet bij de klinische studies. Je enthousiasme voor het

onderzoek, je visie op het leven en je gevoel voor humor zal ik niet snel vergeten. Zonder jouw

inzet hadden we nooit de inclusie voor de ketotifen-trial, nooit binnen de deadline gehaald!

Dr Ankum, beste Pim. Je input bij de klinische studies was van onschatbare waarde. Je

toegankelijkheid en belangeloze inzet waardeer ik enorm. Professor Matthe Burger, Dr. Ko

van der Velden, Dr. Mark van Beurden (je komt toch wel je beloofde biertje innen?). Ik dank

jullie voor de medewerking en het enthousiasme waarmee jullie me geholpen hebben.

Ook de verpleging van H5zuid (ondanks alle bisacodylletjes) en de dames van de poli

gynaecologie wil ik heel erg bedanken voor hun behulpzaamheid en gastvrijheid.

Professor Hollmann, beste Markus en Dr. Hofland, beste Jan. Jullie expertise op het

gebied van de anesthesie was van onschatbare waarde voor het tot stand komen van de

klinische studies. Jullie deur stond altijd open, fantastisch. Ik hoop dat we nog eens wat

leuke projecten samen kunnen gaan opzetten. Mijn dank aan jullie is groot.

Professor Gouma, Dr. Olivier Busch en Professor Willem Bemelman, beste heren. Ondanks

het grote aantal studies dat op de afdeling chirurgie loopt was de behulpzaamheid en

gastvrijheid vanaf het begin groot. In een constructieve sfeer en met de motivatie om

gezamenlijk mooi onderzoek te doen was het altijd mogelijk om een oplossing te zoeken.

Ik dank jullie voor de goede samenwerking en de behulpzaamheid.

De afdeling Maag-, Darm- en Leverziekten wil ik bedanken voor het bieden van een

inspirerende werkplek waar ik van af het eerste moment het gevoel had graag te willen

werken. Professor Bartelsman, beste Joep. Het enthousiasme waarmee jij het vak weet over

te brengen is aanstekelijk en heeft er voor gezorgd dat ik dit vak graag wil uitoefenen.

Beste Robert, mister Apple! De beste “sidekick” die een arts-assistent zich kan wensen!

Leuke en boeiende gesprekken over medische, ethische, maatschappelijke en elektronische

onderwerpen in het OLVG en op de fiets naar huis blijven me bij. Solidair tot in de late

uurtjes. Zonder jou was dit boekwerk letterlijk nooit geworden wat het nu is (een esthetische

aanwinst voor iedere boekenkast)! Ik bewonder je veelzijdigheid en extreem sociale inborst.

Ik hoop dat we elkaar niet uit het oog verliezen, dank!

Page 245: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

244

Gabor en Jesse. Bij het typen van jullie namen laat ik een traan. Jullie steun, zowel

vakinhoudelijk, wetenschappelijk en als vrienden is met geen pen te beschrijven. Jullie

onvoorwaardelijke vriendschap heb ik meer dan eens op de proef gesteld. Jullie stonden

altijd klaar met raad en daad. Zonder jullie hulp had ik dit nooit kunnen volbrengen. De drie

musketiers ride once more!

Zonder vrienden is het leven zinloos! Sanne, Alain, Fanny, Bart, Barbara, Michiel, Petra,

Laurens, Marjolein, Taco, Annet, Martine maar ook al die anderen. Jullie steun, vertrouwen

en relativeringsvermogen waren onontbeerlijk. Ik hoop nog lang en vooral ook vaker van

jullie vriendschap te mogen genieten.

Anne-Mei, Onno, Mey Mey en Ying. Lieve zus en familie, dank voor jullie peptalk en de

heerlijke momenten samen. Ze waren nodig om zo nu en dan weer even inspiratie op te

doen. Bert en Joke, dank voor het in mij gestelde vertrouwen.

Willemijn, de liefde van mijn leven! Ik ben je eeuwig dankbaar voor de onvoorwaardelijke

steun die me gegeven hebt. Je hebt gezorgd voor de basis en de ideale conditie waaronder

ik kon werken. Nooit heb je geklaagd als ik weer in het weekend naar het AMC moest of

wanneer er tijdens de vakantie aan een stuk gewerkt moest worden. Je weet niet half wat

dit voor mij betekend heeft. En dan te bedenken dat je zelf met een prestigieus AGIKO

project bezig bent! Ik hoop dat ik je de komende tijd iets kan teruggeven van alles wat je

mij gegeven hebt

Hauw en Mariet, mijn lieve ouders. Wie had ooit gedacht dat het dromertje dat zich door

zijn school carrière heen moest worstelen ooit nog eens zou promoveren! Zonder jullie

blindelings vertrouwen en steun had ik het in ieder geval nooit gered! Ik ben jullie innig

dankbaar voor alles wat jullie me hebben meegegeven.

Page 246: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

11

Sam

enva

tting

en

conc

lusie

s,

dank

woo

rd e

n co

lour

figu

res

245

Page 247: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

246

Colour figuresg 3

C D

E F

A B

G HChapter 2 - figure 4

Page 248: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

11

Sam

enva

tting

en

conc

lusie

s,

dank

woo

rd e

n co

lour

figu

res

247

A B

C D

200 m

EChapter 3 - figure 6

Page 249: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

248

IM sham IM VNS1V IM VNS5V

IM VNS5V + HexaL VNS5V + Hexa IM VNS5V + vehicle

C

Chapter 4 - figure 6

Page 250: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

11

Sam

enva

tting

en

conc

lusie

s,

dank

woo

rd e

n co

lour

figu

res

249

B

IM VNSIM shamL shamCMLM

SM

MP

PYStat-3 dextran DaPi

C

Amacrophages

Chapter 4 - figure 8

Page 251: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

250

Nicotine

Vehicle

AR-R17779

3TATS-YP & 08/4F egrem3TATS-YP08/4F

B

Chapter 5 - figure 4

Page 252: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

11

Sam

enva

tting

en

conc

lusie

s,

dank

woo

rd e

n co

lour

figu

res

251

A B

DC

Chapter 7 - figure 3

Page 253: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

252

Chapter 7 - figure 5

A B

Kit/WT

Kit/Kit v

Kit/WTMC

Kit/Kit v

PBS

Kit/Kit v

A B

C D

Chapter 7 - figure 9

Page 254: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

11

Sam

enva

tting

en

conc

lusie

s,

dank

woo

rd e

n co

lour

figu

res

253

Chapter 7 - figure 7

LM CM

PP

LM

CM

A

C

B

D

E F

Page 255: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

254

20.00 m

A

Chapter 8 - figure 1

Page 256: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

11

Sam

enva

tting

en

conc

lusie

s,

dank

woo

rd e

n co

lour

figu

res

255

4

preoperative scan postoperative scan

Chapter 8 - figure 4

Page 257: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

256

rolling

bacterialtranslocation

adhesionactivation

ICAM-1 antibody/antisenseICAM-1

diapedesis

A

LFA-1

C

B

E

D

G

F

mast cell stabilizers

macrophage activation

vagus nerve

generalized hypomotilityi.e. POSTOPERATIVE ILEUS

electrical stimulation

Inhibitory neuralpathway

activation

Chapter 10 - Summarizing figure

Page 258: Pathophysiology of Postoperative - Lygature of Postoperative Ileus: from Bench to Bedside ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor aan …

Cha

pter

11

Sam

enva

tting

en

conc

lusie

s,

dank

woo

rd e

n co

lour

figu

res

257