SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

131
Sphingosine-1-phosphate effects on conventional outflow physiology Item type text; Electronic Dissertation Authors Sumida, Grant Publisher The University of Arizona. Rights Copyright © is held by the author. Digital access to this material is made possible by the University Libraries, University of Arizona. Further transmission, reproduction or presentation (such as public display or performance) of protected items is prohibited except with permission of the author. Downloaded 18-Feb-2018 02:24:26 Link to item http://hdl.handle.net/10150/194893

Transcript of SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

Page 1: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

Sphingosine-1-phosphate effects on conventional outflowphysiology

Item type text; Electronic Dissertation

Authors Sumida, Grant

Publisher The University of Arizona.

Rights Copyright © is held by the author. Digital access to thismaterial is made possible by the University Libraries,University of Arizona. Further transmission, reproductionor presentation (such as public display or performance) ofprotected items is prohibited except with permission of theauthor.

Downloaded 18-Feb-2018 02:24:26

Link to item http://hdl.handle.net/10150/194893

Page 2: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

 

SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL OUTFLOW PHYSIOLOGY

by

Grant Masaaki Sumida

_____________________

A Dissertation Submitted to the Faculty of the

GRADUATE INTERDISCIPLINARY PROGRAM IN PHYSIOLOGICAL SCIENCES

In Partial Fulfillment of the Requirements For the Degree of

DOCTOR OF PHILOSOPHY

In the Graduate College

THE UNIVERSITY OF ARIZONA

2010

Page 3: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

 

THE UNIVERSITY OF ARIZONA GRADUATE COLLEGE

As members of the Dissertation Committee, we certify that we have read the dissertation prepared by Grant Masaaki Sumida entitled Sphingosine-1-phosphate effects on conventional outflow physiology and recommend that it be accepted as fulfilling the dissertation requirement for the Degree of Doctor of Philosophy _______________________________________________________________________ Date: 10/29/2010

W. Daniel Stamer, Ph.D. _______________________________________________________________________ Date: 10/29/2010

Ronald Lynch, Ph.D. _______________________________________________________________________ Date: 10/29/2010

Scott Boitano, Ph.D. _______________________________________________________________________ Date: 10/29/2010

Nicholas Delamere, Ph.D. _______________________________________________________________________ Date: 10/29/2010

Josephine Lai, Ph.D. Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation to the Graduate College. I hereby certify that I have read this dissertation prepared under my direction and recommend that it be accepted as fulfilling the dissertation requirement. ________________________________________________ Date: 10/29/2010 Dissertation Director: W. Daniel Stamer, Ph.D.

Page 4: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

 

STATEMENT BY AUTHOR This dissertation has been submitted in partial fulfillment of requirements for an advanced degree at the University of Arizona and is deposited in the University Library to be made available to borrowers under rules of the Library. Brief quotations from this dissertation are allowable without special permission, provided that accurate acknowledgment of source is made. Requests for permission for extended quotation from or reproduction of this manuscript in whole or in part may be granted by the head of the major department or the Dean of the Graduate College when in his or her judgment the proposed use of the material is in the interests of scholarship. In all other instances, however, permission must be obtained from the author. SIGNED: Grant Masaaki Sumida

Page 5: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

 

ACKNOWLEDGEMENTS

I would like to thank my advisor, Dr. Daniel Stamer for guiding me in my development as a researcher. Also, to the rest of my dissertation committee, Dr. Ronald Lynch, Dr. Scott Boitano, Dr. Nicholas Delamere, and Dr. Josephine Lai for their helpful input, criticism, and encouragement throughout my dissertation research.

I also appreciate everyone in the Stamer lab for teaching me not only how to conduct research, but also how to conduct myself through life. I especially would like to thank Dr. Renata Ramos and Dr. Nicholas Baetz for helping me through my first years of graduate school. I also would like to thank Emely Hoffman and Krisitin Perkumas for supplying cells, ordering reagents, and their stabilizing presence.

I would also like to thank our collaborators, Dr. C. Ross Ethier for his helpful discussions and Dr. Darryl Overby for introducing the whole-eye perfusion system to our laboratory. Also, my special thanks to the tissue banks: Sun Health Research Institute, National Disease Research Interchange, and Life Legacy Foundation. I must also acknowledge the outstanding volunteering effort put forth by the Lion’s Club in transporting tissue.

Finally, I would like to thank my family and friends for the unwavering support they have provided me for all these years.

This work was supported by the National Institute of Health and the Research to Prevent Blindness Foundation.

Page 6: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

 

DEDICATION

To my family.

And Bernadette.

Page 7: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

 

TABLE OF CONTENTS

LIST OF FIGURES ......................................................................................... 9 LIST OF TABLES ......................................................................................... 11 ABSTRACT .................................................................................................. 12 1. INTRODUCTION, HYPOTHESIS, AND SPECIFIC AIMS ................. 14 1.1 Introduction .............................................................................................................14 1.2 Hypothesis ...............................................................................................................18 1.3 Specific aims ...........................................................................................................18 2. CRITICAL LITERATURE REVIEW ..................................................... 19 2.1 Glaucoma.................................................................................................................19 2.1.1 Primary open-angle glaucoma .........................................................................19 2.1.2 Loss of retinal ganglion cells ..........................................................................20 2.2 Generation of intraocular pressure ..........................................................................21 2.2.1 Aqueous humor secretion ................................................................................23 2.2.2 Aqueous humor outflow ..................................................................................24 2.3 Conventional outflow pathway ...............................................................................24 2.3.1 Trabecular meshwork ......................................................................................25 2.3.2 Schlemm’s canal .............................................................................................26 2.3.3 Cytoskeletal network in the cells of the outflow pathway ..............................28 2.4 Sphingosine-1-phosphate receptor signaling...........................................................29 2.4.1 Formation and export of sphingosine-1-phosphate .........................................29 2.4.2 Sphingosine-1-phosphate receptors .................................................................30

2.4.3 Sphingosine-1-phosphate effects on the cytoskeleton and junction assembly ............................................................................................32

2.4.4 Sphingosine-1-phosphate receptor compounds ...............................................35 2.5 Sphingosine-1-phosphate receptor signaling and the conventional outflow pathway ......................................................................................................38 3. SPHINGOSINE-1-PHOSPHATE EFFECTS ON THE INNER WALL OF SCHLEMM’S CANAL AND OUTFLOW FACILITY IN PERFUSED HUMAN EYES................................................ 39 3.1 Introduction .............................................................................................................39 3.2 Methods ...................................................................................................................42 3.2.1 Cell culture ......................................................................................................42 3.2.2 Reverse-transcription polymerase chain reaction............................................42

Page 8: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

 

TABLE OF CONTENTS – Continued

3.2.3 Western blot analyses ......................................................................................43 3.2.4 Immunofluorescence microscopy of ocular sections ......................................44 3.2.5 Whole-eye perfusion and facility measurements ............................................45 3.2.6 Labeling and confocal microscopy of the inner wall ......................................48 3.2.7 SEM and semi-thin sectioning ........................................................................49 3.3 Results .....................................................................................................................51 3.4 Discussion ...............................................................................................................59 4. SPHINGOSINE-1-PHOSPHATE ENHANCEMENT OF CORTICAL ACTOMYOSIN ORGANIZATION IN CULTURED HUMAN SCHLEMM’S CANAL ENDOTHELIAL CELL MONOLAYERS ................................................... 63 4.1 Introduction .............................................................................................................63 4.2 Methods ...................................................................................................................66 4.2.1 Cell culture ......................................................................................................66 4.2.2 Immunoblot analyses .......................................................................................66 4.2.3 RhoA activation assay .....................................................................................67 4.2.4 Rac activation assay ........................................................................................68 4.2.5 Immunofluorescence microscopy ...................................................................69 4.3 Results .....................................................................................................................71 4.4 Discussion ...............................................................................................................79 5. S1P2 RECEPTOR REGULATION OF SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL OUTFLOW PHYSIOLOGY… .......................................................................................... 83 5.1 Introduction .............................................................................................................83 5.2 Methods ...................................................................................................................85 5.2.1 Reagents ..........................................................................................................85 5.2.2 Eye tissue .........................................................................................................85 5.2.3 Cell culture ......................................................................................................85 5.2.4 Immunoblot analyses .......................................................................................86 5.2.5 Human whole-eye perfusion ...........................................................................87 5.2.6 Porcine whole-eye perfusion ...........................................................................88 5.2.7 Statistical analyses ...........................................................................................88 5.3 Results .....................................................................................................................90 5.4 Discussion .............................................................................................................100 6. CONCLUSIONS .....................................................................................104 6.1 Summary ...............................................................................................................104

Page 9: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

 

TABLE OF CONTENTS - Continued 6.2 Interpretation .........................................................................................................106 6.3 Future direction .....................................................................................................109 6.3.1 Sphingosine-1-phosphate perfusion ..............................................................109 6.3.2 SC cell model ................................................................................................111 6.4 Perspective.............................................................................................................113 APPENDIX A – WHOLE-EYE PERFUSION MODEL .............................114 REFERENCES .............................................................................................117

Page 10: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

 

LIST OF FIGURES

Figure 1.1 Diagram of the human eye .........................................................................15 Figure 2.1 Aqueous humor flow..................................................................................22 Figure 2.2 Conventional outflow pathway ..................................................................25 Figure 2.3 MLC phosphorylation and cell contractility through RhoA

activation ....................................................................................................34 Figure 2.4 Chemical structures of subtype-specific S1P receptor

compounds .................................................................................................37 Figure 3.1 Western blot analyses of S1P receptor subtype expression

in Schlemm’s canal endothelial cells .........................................................52 Figure 3.2 Expression of S1P receptor subtypes by human Schlemm’s

canal endothelia in situ ...............................................................................53 Figure 3.3 Effect of S1P on outflow facility in paired whole human

eye perfusions ............................................................................................54 Figure 3.4 Morphological analyses of the inner wall of Schlemm’s

canal and associated conventional outflow structures in human eyes perfused with S1P ..................................................................55

Figure 3.5 Actin architecture and Vascular Endothelial (VE)-cadherin

expression in Schlemm’s canal inner wall of eyes perfused with S1P .....................................................................................................56

Figure 3.6 Actin architecture and β-catenin expression in Schlemm’s

canal inner wall of eyes perfused with S1P ...............................................58 Figure 4.1 S1P-induced RhoA activation and MLC phosphorylation in

SC cell monolayers ....................................................................................71 Figure 4.2 Dose-dependent S1P-mediated MLC phosphorylation in

both TM and SC cell monolayers ..............................................................72 Figure 4.3 Rho-kinase-dependent MLC phosphorylation by S1P in SC

cell monolayers ..........................................................................................73

Page 11: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

10 

 

LIST OF FIGURES - Continued

Figure 4.4 MLC expression levels in TM and SC cell monolayers ............................74 Figure 4.5 S1P-induced Rac activation in SC cell monolayers ...................................75 Figure 4.6 S1P promotes cortical actin assembly and peripheral

localization of phospho-MLC in SC cell monolayers................................76 Figure 4.7 S1P and phospho-MLC localization in HUVEC .......................................77 Figure 4.8 S1P and β-catenin localization in SC cells ................................................77 Figure 4.9 Model for differential S1P receptor coupling to Rac/Rho

pathways in outflow cells...........................................................................81 Figure 5.1 S1P1 receptor activation does not promote MLC

phosphorylation..........................................................................................91 Figure 5.2 Effect of a single concentration of S1P receptor specific

antagonist on MLC phosphorylation in the presence of increasing concentrations of S1P ...............................................................93

Figure 5.3 Effect of increasing antagonist concentration on S1P-

mediated MLC phosphorylation in SC ......................................................94 Figure 5.4 Effect of increasing antagonist concentration on S1P-

mediated MLC phosphorylation in TM cells .............................................95 Figure 5.5 Blocking S1P2 prevents the S1P-mediated decrease in

outflow facility in porcine eyes ..................................................................97 Figure 5.6 Outflow facility effects of S1P2 receptor blockade in S1P-

perfused human eyes ..................................................................................99

Page 12: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

11 

 

LIST OF TABLES

Table 3.1 Summary of S1P perfusion results and donor information ........................46 Table 3.2 mRNA expression of S1P receptors in SC and TM cells ..........................52 Table 5.1 Donor information and summary of results from human

whole-eye perfusions .................................................................................90

Page 13: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

12 

 

ABSTRACT

Glaucoma is the leading cause of irreversible blindness worldwide with the most

prevalent form, primary open-angle glaucoma (POAG), accounting for the vast majority

of glaucoma cases. The main risk-factor for POAG is an elevated intraocular pressure

(IOP), and is due to an increased resistance to aqueous humor outflow in the conventional

outflow pathway at the juxtacanalicular region of the trabecular meshwork (TM) and the

inner wall of Schlemm’s canal (SC). Reducing elevated IOP is the most effective method

to prevent further loss of vision in glaucoma; therefore, it is important to understand how

outflow resistance is regulated in the conventional outflow pathway in order to find

effective methods to reduce ocular hypertension.

Sphingosine-1-phosphate (S1P) is an endogenous lipid that reduces outflow

facility in porcine eyes, thereby increasing resistance. S1P plays a major role in affecting

cell migration, endothelial permeability, and junctional formation, processes that are

intimately linked and regulated by cytoskeletal dynamics. Due to S1P’s known effect of

decreasing endothelial permeability in vascular endothelial cells, the overall hypothesis

of this dissertation is that the S1P-induced decrease in outflow facility occurs through a

mechanism that involves S1P receptor activation in SC cells.

The results from the studies within this dissertation demonstrate the expression of

the S1P1-3 receptor subtypes in SC and TM cells and a decrease of outflow facility by S1P

in perfused human eyes. Additionally, S1P promotes F-actin formation and myosin light

chain (MLC) phosphorylation at the SC cell cortex. The S1P-promoted MLC

phosphorylation in both SC and TM cells, in addition to the S1P-induced decrease of

Page 14: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

13 

 

outflow facility in porcine and human eyes, were blocked by the S1P2 antagonist JTE-

013.

Results from these studies demonstrate S1P to actively regulate actomyosin

dynamics in the cells of the outflow pathway through the S1P2 receptor. S1P2 also

mediates the S1P-induced increase in outflow resistance. Therefore, S1P2 is a novel

pharmacological target in the conventional outflow pathway to reduce elevated IOP in

glaucoma patients.

 

Page 15: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

14 

 

1. INTRODUCTION, HYPOTHESIS, AND SPECIFIC AIMS

1.1 Introduction

Glaucoma is the leading cause of irreversible blindness worldwide and the second

leading cause of blindness in the United States. There are an estimated 60.5 million

people with glaucoma today and 79.6 million people are predicted to have glaucoma by

2020 [1]. Glaucoma is defined by structural and functional abnormalities in the eye with

the observation of optic neuropathy and a visual field defect, respectively [2]. The

resultant loss of vision from glaucoma is attributed to the loss of retinal ganglion cells

and their axons at the optic nerve head [3, 4]. Of the various forms of glaucoma, primary

open-angle glaucoma (POAG) is the most prevalent, accounting for more than 90% total.

Glaucoma is categorized as POAG when the angle formed between the cornea and iris

remains open, and appears normal with no other physical obstruction or morphological

changes apparent (refer to Figure 1.1 for eye anatomy).

One of the primary risk-factors associated with POAG is an elevated intraocular

pressure (IOP). Elevated IOP is of particular importance in glaucoma because unlike

other glaucoma risk-factors (ex. age, race, family history), elevated IOP is modifiable.

IOP is generated by a balance between aqueous humor secretion by the ciliary epithelium

and aqueous humor outflow, but IOP is entirely regulated by the resistance to outflow.

The majority (70-90%) of aqueous humor exits through the conventional outflow

pathway, consisting of the trabecular meshwork (TM) and Schlemm’s canal (SC).

Within the conventional outflow pathway, the majority (~90%) of the resistance to

aqueous humor outflow is commonly thought to be located in the juxtacanalicular

Page 16: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

15 

 

Figure 1.1. Diagram of the human eye. Courtesy: National Eye Institute, National Institutes of Health.

connective tissue region of the TM and the inner wall of SC [5-8]. Although the location

of the majority of outflow resistance is known, the precise molecular mechanism

regulating resistance is not. Therefore, it is essential to both determine that molecular

mechanism and identify new pharmacological targets to reduce outflow resistance and

IOP in glaucoma patients.

In searching for new compounds and targets to affect outflow resistance, the

bioactive lipid sphingosine-1-phosphate (S1P) was found to induce a significant decrease

in outflow facility (increase in outflow resistance) in the porcine whole-eye perfusion

model [9]. Additionally, S1P was found to increase stress fiber formation and myosin

light chain phosphorylation in cultured TM cells [9], suggesting that contraction of the

TM increases resistance by decreasing available flow pathways for aqueous humor

through the TM. The significant and sustained decrease of outflow facility by S1P in the

Page 17: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

16 

 

porcine perfusion model has identified S1P and its downstream signaling in TM cells as a

prominent pathway in regulating outflow resistance.

S1P is an endogenous lipid originally thought to only be an intermediate step in

lipid synthesis and breakdown. Following the identification of S1P as the ligand for the

G-protein coupled receptor S1P1 [10], interest in S1P, its receptors, and downstream

signaling following S1P receptor activation have increased. S1P binds with high affinity

to 5 receptor subtypes (S1P1-5) and is now known to play a key role in many processes

such as bradycardia, vasoconstriction, angiogenesis, and lymphocyte egress [11, 12]. At

the cellular level, S1P affects cytoskeleton dynamics and plays a role in processes such as

cell migration and endothelial barrier regulation [13]. In particular, S1P-treated

endothelial cells decrease endothelial permeability by increasing junctional proteins to

sites of cell-cell contact [14-16]. Additionally, S1P promotes actomyosin organization at

the cell cortex to stabilize the cell-cell and cell-matrix junctions [17-19].

S1P has been shown to increase outflow resistance in the porcine perfusion model

and affect TM cell contractility [9], but the effects of S1P on outflow in humans has not

been investigated, nor the effects of S1P on the cells of the inner wall endothelia. Is the

effect of S1P on the human conventional outflow pathway similar to that observed in the

porcine model, a species with a significantly different outflow apparatus? Does S1P

affect the cells of the inner wall of SC, and if so, how would they contribute to an overall

change in outflow resistance? As a continuous endothelial monolayer with similarities to

vascular endothelia [20], S1P may decrease endothelial permeability across the inner wall

of SC by promoting an increase in junctional organization. In support of a decrease in

Page 18: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

17 

 

endothelial permeability across the inner wall, histology of S1P-treated porcine eyes

displayed increased giant vacuole formations along the endothelial lining of the aqueous

plexus (analogous to SC) compared to control, indicative of an increased pressure-drop

(as well as a higher resistance) across the endothelial lining [9]. Lastly, will affecting the

S1P receptor system be a potential pharmacological target to help reduce ocular

hypertension? With the expression of multiple S1P receptor subtypes throughout the

body, it is important to determine if the S1P effect on outflow resistance is mediated by a

specific subtype, or multiple subtypes. Subsequent pharmacological targeting of the

specific receptor(s) may prove to be an effective method in decreasing outflow resistance

in the conventional outflow pathway.

The objective of this dissertation project is to provide answers for these questions

with the goal of contributing to our general understanding of how outflow is regulated

and to identify a potential target for glaucoma therapeutics.

Page 19: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

18 

 

1.2 Hypothesis

S1P decreases outflow facility of perfused human eyes in organ-culture by a

mechanism that involves S1P receptor activation in Schlemm’s canal cells.

1.3 Specific aims

To test the central hypothesis, I formulated three specific aims. Specific aim 1

was to characterize the expression of S1P receptors in SC endothelial cells. I

hypothesized that the S1P1 and S1P3 receptors are expressed by the SC endothelial cells.

Specific aim 2 was to identify the downstream effectors of S1P receptor activation

in SC endothelial cells. I hypothesized that SC cells in culture respond to S1P by

increasing actomyosin organization at the cell cortex and promoting junctional

organization.

Specific aim 3 was to identify the S1P receptors responsible for S1P’s effect in

human whole-eye organ culture. I hypothesized that activation of the S1P1 receptor in

the conventional outflow pathway contributes to the S1P-induced decrease in outflow

facility.

Page 20: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

19 

 

2. CRITICAL LITERATURE REVIEW

2.1 Glaucoma

Glaucoma is the second leading cause of blindness behind cataract and the leading

cause of irreversible blindness in the world. Clinically, glaucoma presents as an optic

neuropathy with the loss of retinal ganglion cells (RGC) and their axons at the optic

nerve head [2]. The characteristic increase in the cup:disc ratio at the optic nerve head of

this neurodegenerative disease can be directly observed by ophthalmoscopy and is a

result of a thinning of the layer of nerve fibers (loss of RGC axons) running through to

the optic nerve and the compression of the lamina cribrosa with subsequent deepening of

the optic nerve head [21]. Glaucoma is functionally presented as a loss of vision (mid-

peripheral first) that is assessed through a visual field test [2]. There are different

conditions leading to the various forms of glaucoma, giving rise to the common

characteristic loss of RGCs at the optic nerve head leading to a decreased visual field. Of

the various forms of glaucoma, the most common is primary open-angle glaucoma

(POAG).

2.1.1 Primary open-angle glaucoma

In POAG, the open-angle refers to the angle formed by the cornea and iris; a

closed-angle occurs with displacement of the iris anteriorly with the subsequent blocking

of the trabecular meshwork (discussed in chapter 2.3.1). Primary in POAG refers to the

appearance of the disease without other observable factors or conditions that may be

responsible or contribute to the development of glaucoma. Due to the slow, progressive

Page 21: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

20 

 

nature of the disease with the loss of peripheral vision first, an estimated 50% of people

with open-angle glaucoma in developed countries are undiagnosed [22].

There are many risk-factors strongly associated with POAG. African-Americans

were observed to be at higher risk for POAG than whites [23] while increasing age is

another risk-factor regardless of race [24]. Family history is a also a strong risk-factor for

POAG, with relatives of patients with glaucoma at a higher risk for the disease [25].

With family history as a risk-factor for POAG, the search for defected ‘glaucoma genes’

has been underway with several genes already identified [26].

Of the risk-factors strongly associated with POAG, only elevated IOP is

modifiable and therefore, treatable. An increased IOP is thought to contribute to

glaucoma by blocking axonal transport in RGCs at the optic nerve head at the level of the

lamina cribrosa, thus leading to optic neuropathy (see 2.1.2). Elevated IOP (≥ 21

mmHg), originally included in the definition of glaucoma, is a strong factor in the disease

but is not a requirement for the onset of glaucoma due to the occurrence of POAG in

patients with IOP in the normal range [23, 27, 28]. The occurrence of POAG at varying

levels of IOP suggests differences in susceptibility to RGC damage from IOP between

individuals and hints at other contributing factors yet to be determined [29].

2.1.2 Loss of retinal ganglion cells

The cone and rod photoreceptors capture light in the posterior portion of the

retina. After a series of connections between various neuroretinal cells (bipolar,

amacrine, and horizontal cells) that modulate and organize the information gathered by

Page 22: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

21 

 

the photoreceptors, the visual information is conducted by the long axons of RGCs from

the retina to the lateral geniculate body and other processing centers in the brain.

Approximately 1 million RGC axons run along the posterior wall of the eye and converge

to form the optic nerve. Bundles of axons must pass through the lamina cribrosa, an

opening in the scleral wall consisting of sieve-like laminar plates of connective tissue.

Glaucoma is characterized by a progressive loss of RGCs and their axons at the

optic nerve head. The site of retinal ganglion cell injury was demonstrated in monkeys

with experimentally raised IOP; the block of axonal transport in RGCs, both orthograde

and retrograde, was observed at the optic nerve head [30-32]. Additionally, the DBA/2J

glaucoma mouse model also demonstrates progressive axonal damage of RGCs at the

level of the glial lamina (analogous to lamina cribrosa) with increased IOP [33]. The

glaucomatous phenotype in the DBA/2J animal is due to a pigment dispersion disease of

the iris that results in blockage of the outflow pathway and an increase in IOP, resulting

in optic neuropathy with increasing age. In post-mortem, enucleated human eyes from

glaucomatous donors, damage of RGC axons were observed at the same locus [4]. The

close resemblances of RGC damage in glaucoma patients to the damage induced

experimentally by raising IOP in animal models, again, shows a link between POAG and

susceptibility of RGCs and their axons to injury with increased pressure.

2.2 Generation of intraocular pressure

IOP is generated by a balance between the secretion of aqueous humor by the

ciliary epithelium, and its drainage through the outflow pathways (Figure 2.1). Although

Page 23: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

22 

 

an elevated IOP is not a requirement for POAG [28], it is still an important factor in the

development of the disease. To highlight the relationship between POAG and high IOP,

glaucoma patients that maintained a low IOP following surgery to increase outflow did

not demonstrate further visual field loss compared to the patients that did not maintain a

low IOP [34]. Therefore, the regulation of elevated IOP, either by reducing aqueous

humor secretion or increasing its outflow, is at the forefront for glaucoma therapeutics.

Figure 2.1. Aqueous humor flow. Aqueous humor is constantly secreted by the ciliary epithelium lining the ciliary body. The majority of the aqueous humor exits through the conventional outflow pathway that sits at the angle formed by the cornea and iris and consists of the trabecular meshwork and Schlemm’s canal. Courtesy: National Eye Institute, National Institutes of Health.

Page 24: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

23 

 

2.2.1 Aqueous humor secretion

Aqueous humor is a clear, colorless liquid that is continually secreted by the

ciliary epithelium lining the ciliary body and drained through the outflow pathways. The

flow rate (secretion) of aqueous humor averaged over 24 hrs is 2.3 µl/min; the average

rate of flow during the daytime (0800-1600 hrs) is 2.8 µl/min [35]. Besides providing a

clear colorless medium through which light may pass through, aqueous humor also serves

an important role in maintaining proper IOP and providing nutrients and removing waste

for the avascular tissues of the eye (cornea, trabecular meshwork, and lens).

Secretion of aqueous humor into the posterior chamber is based on the active

transport of solutes by the ciliary epithelium. The ciliary epithelium consists of two

epithelial layers connected at their apical membranes and in direct communication with

one another through gap junctions, thus allowing the two layers to function as a unit. The

pigmented epithelial cell layer faces the stroma of the ciliary processes while the non-

pigmented epithelial layer faces the posterior chamber. A general net transfer of NaCl

from the stroma to the posterior chamber occurs in three steps: 1) NaCl enters the

pigmented epithelium from the stroma, 2) solutes cross over to the non-pigmented

epithelium via gap junctions, and 3) Na+ is pumped into the posterior chamber (with Cl-

following through Cl- channels) [36]. As solutes are actively pumped into the posterior

chamber, water follows.

Page 25: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

24 

 

2.2.2 Aqueous humor outflow

Once aqueous humor is secreted into the posterior chamber, it flows through the

pupil and into the anterior chamber. The majority of aqueous humor exits through the

conventional outflow pathway, consisting of the trabecular meshwork (TM) and

Schlemm’s canal (SC). Aqueous humor may also exit through the uveoscleral pathway

located at the iris root by entering the interstitial spaces of the ciliary muscle [37]. In

humans, the outflow through the uveoscleral pathway accounts for less than 20% of total

outflow [38]. The uveoscleral pathway, unlike the conventional outflow pathway, is

pressure-independent [39] and flow through this pathway decreases with age [40].

Along with handling the majority of the aqueous humor outflow, the conventional

outflow pathway is also responsible for generating the majority of resistance to outflow

[41]. It is this resistance to outflow that regulates IOP levels. Additionally, the resistance

to outflow increases with age and POAG [42]. Within the conventional outflow pathway,

the majority of resistance is located at or near the juxtacanalicular connective tissue

(JCT) region of the TM and inner wall of SC [5-8]. The increased resistance to outflow

at the JCT and inner wall region leads to an elevated IOP and therefore, is a target in

understanding how outflow and the generation of IOP are regulated.

2.3 Conventional outflow pathway

The majority of aqueous humor in the anterior chamber is returned to venous

circulation via the conventional outflow pathway that lies near the angle formed by the

cornea and iris (Figure 2.2). After aqueous humor crosses the TM and inner wall of SC,

Page 26: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

25 

 

it enters collector channels and aqueous veins before returning to venous circulation. The

conventional outflow pathway is pressure-dependent with the outflow rate of aqueous

humor increasing linearly with increasing pressure.

Figure 2.2. Conventional outflow pathway. The majority of aqueous humor exits the anterior chamber (AC) through the conventional outflow pathway, consisting of the trabecular meshwork (TM) and Schlemm’s canal (SC). Arrow indicates flow of aqueous humor. CC: collector channel. Photo taken by Renata Fortuna Ramos.

2.3.1 Trabecular meshwork

The TM comprises three layers, the uveal, corneoscleral, and JCT. The first two

layers that aqueous humor encounters, the uveal and corneoscleral meshwork, consists of

a series of trabecular beams covered by a single layer of TM cells. The trabecular beams,

arranged in 12-20 layers with 3-5 layers at the anterior TM, forms the flow channels that

aqueous humor must first pass through upon exiting the anterior chamber [43]. The

Page 27: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

26 

 

space between the trabecular beams in the corneoscleral meshwork get progressively

smaller as the aqueous humor flows from the inner to outer regions of the TM. The TM

cells are phagocytic and provide a valuable role in clearing material such as pigment and

preventing the occlusion of aqueous outflow [44-46]. But with increasing age, there is a

loss of TM cells with few replacements, thus resulting in an overall decrease of TM cells

[47].

The third and final layer of the TM is the JCT layer, located adjacent to the inner

wall of SC. Unlike the uveal and corneoscleral layers that contain trabecular beams

covered by a single TM cell layer, the JCT is a 2-20 µm thick region that comprises a

loose arrangement of extracellular matrix with TM cells arranged in 2-5 layers [43]. The

TM cells form attachments to neighboring TM cells through long processes, as well as

attachments to the underlying matrix and endothelial cells of the inner wall. Aqueous

humor flows through the space left unoccupied by cells and the ECM.

2.3.2 Schlemm’s canal

Aqueous humor returns to venous circulation by entering SC, a lumen located

within the scleral sulcus and encircling the anterior segment of the eye. The lumen of SC

is also continuous with around 30 collector channels, leading to the episcleral veins. The

side of SC that is adjacent to the sclera is the outer wall while the opposite side adjacent

to the TM is the inner wall.

The inner wall of SC comprises elongated endothelial cells 100-150 µm long that,

as opposed to the flat outer wall, appears highly irregular with a flat to undulating

Page 28: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

27 

 

appearance [43]. This appearance of the inner wall may be related to the segmental flow

of aqueous humor, crossing into SC at various locations in a non-uniform fashion [48].

The inner wall is a continuous endothelial monolayer on a discontinuous basal lamina

that aqueous humor must pass in a basal to apical direction [49].

For aqueous humor to cross the inner wall, two pathways have been described, the

trans-cellular and paracellular pathways. Although, the presence of tight junctions in SC

cells may restrict the amount of paracellular flow of aqueous humor crossing between SC

cells [50]. Aqueous humor is thought to cross into SC through pores found throughout

the inner wall [8] with intracellular pores for the intracellular route and border pores for

the paracellular route [51]. A relationship between pore density on the inner wall and

outflow facility was found [52]; unfortunately, the number of pores are unknown due to

fixation conditions (volume of fixative perfused) affecting the number of pores observed.

It is currently estimated that there are less than 1000 pores/mm2 in a normal eye at any

one instant [51, 53].

Another feature of the inner wall is the appearance of giant vacuoles, structures

formed by the separation of the inner wall endothelium from the underlying basement

membrane and protruding into SC [49, 54]. When originally observed, the structures

were described as vacuole-like [55]; subsequent experiments using a ferritin tracer

solution demonstrated that the vacuoles are exposed to and filled with aqueous humor

[56]. Giant vacuole formation is thought to be pressure-dependent, with the numbers

and size of the vacuoles directly related to IOP [57]. Giant vacuoles have a short survival

time (< 3 min), therefore, it is believed that they are able to respond rapidly to transient

Page 29: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

28 

 

changes in IOP [58]. Interestingly, a greater number of giant vacuoles are observed near

collector channels where aqueous humor flow is greatest and a larger trans-endothelial

pressure gradient is located [59]. Giant vacuoles are also the site of intracellular pores

that provide routes for aqueous humor to exit into SC [56, 60]. Although pore formations

often occur through giant vacuoles, it appears their formation may be flow-dependent,

rather than pressure-dependent like the giant vacuoles [54].

2.3.3 Cytoskeletal network in the cells of the outflow pathway

Cytoskeletal dynamics in the cells of the outflow pathway play a vital role in

maintaining and regulating outflow resistance. In particular, it is well established that

disruption of the actomyosin network decreases resistance to aqueous humor outflow

[61]. For example, the globular (G)-actin stabilizing drug, latrunculin [62], and the F-

actin depolymerizing drug cytochalasin [63, 64], both increase outflow facility, thereby

decreasing resistance, by decreasing filamentous (F)-actin. Similarly, the Rho-kinase

inhibitor Y-27632 [65-67] and the myosin light chain kinase inhibitor ML-7 [68], also

increase outflow facility by disrupting the cell contractile mechanism. The cytoskeleton

provides stability for the cells at cell–cell and cell-matrix adhesions that is vital in

maintaining tissue structure and resistance in the face of varying levels of IOP and flow.

When the inner wall of SC is exposed to a large basal to apical pressure-drop across the

endothelia (with subsequent giant vacuole formation), a strong cytoskeletal network is

required to help anchor the cell and matrix adhesion proteins. Also, the TM can

Page 30: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

29 

 

modulate its contractile tone to increase or decrease aqueous flow patterns depending on

IOP and flow.

2.4 Sphingosine-1-phosphate receptor signaling

Sphingosine-1-phosphate (S1P) is an endogenous lipid that plays a role in many

biological processes, such as bradycardia, vasoconstriction, angiogenesis, and

lymphocyte egress [11, 12]. Initial studies on S1P as a signaling molecule were

performed in vascular research where S1P measurements in human plasma and serum are

in the 10-7 M range, although the majority is bound to components of the plasma and

serum, such as lipoproteins [69, 70]. Since the identification of the endothelial

differentiation gene (EDG) receptor-1 [71] and S1P as its ligand [10], interest in S1P

receptor signaling has increased due to the ubiquitous expression of S1P receptors

throughout the body and the identification of signaling pathways affecting multiple

cellular processes.

2.4.1 Formation and export of sphingosine-1-phosphate

S1P is synthesized from ceramide, a molecule that provides the initial structure

for the majority of sphingolipids. There are two ways for ceramide to be produced, either

the de novo pathway that occurs on the cytoplasmic surface of the endoplasmic reticulum

and is initiated with the condensation of serine and palmitate by serine

palmitoyltransferase, or through the hydrolysis of complex lipids [72]. Ceramidases

catalyze the formation of sphingosine from ceramide, and S1P may then be formed by the

Page 31: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

30 

 

addition of a phosphate group that is catalyzed by sphingosine kinase (SK) 1/2. S1P may

then be converted back to sphingosine by S1P phosphatase, or irreversibly broken down

into ethanolamine phosphate and hexadecenal by S1P lyase.

S1P is mainly made on the inner leaflet of the plasma membrane [73, 74], but yet,

is present in the extracellular space and is a highly specific ligand to a group of S1P

receptors. There are thought to be two mechanisms to generate extracellular S1P. First,

SK1 released by endothelial cells may phosphorylate extracellular sphingosine [75].

Second, S1P may be released by cells into the extracellular space by transporters, with

members of the ATP-binding cassette (ABC) transporter superfamily as prime

candidates. Initially, the ABCC1 transporter was found to export S1P from mast cells

[76], although a subsequent study found ABCC1 to likely not be the ABC transporter

promoting S1P export [77]. Different studies though, have found other members of the

ABC transporter superfamily to be the potential S1P transporter(s) [77-79].

2.4.2 Sphingosine-1-phosphate receptors

Interest in S1P as a potential extracellular signaling molecule increased with the

identification of S1P as the ligand for a functional receptor, EDG1 [10], inducing an

increase in intracellular calcium and inhibiting cAMP formation upon receptor activation

[80]. EDG1 was originally identified through mRNA transcripts as an orphan G protein-

coupled receptor (GPCR) expressed by vascular endothelial cells [71]. Currently, S1P is

known to bind with high affinity to 5 different GPCR subtypes (S1P1-5), formally known

as EDG receptors [81]. Of the 5 S1P receptor subtypes, the S1P1-3 receptors are the most

Page 32: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

31 

 

prevalent with mRNA expression detected throughout the body, while S1P4 expression is

limited to lymphoid and S1P5 to brain and skin [12].

Upon the binding of a ligand to its GPCR, an associated heterotrimeric G protein

becomes activated and capable of inducing a downstream signaling cascade. The S1P1-3

receptors differentially couple to various G proteins; S1P1 couples solely to Gi, while

S1P2 and S1P3 couple to Gi/o, Gq, and G12/13 [82, 83], although S1P2 couples particularly

strong to G12/13.

Initial research on S1P1 has identified this receptor (as well as S1P) as a key

player in vascular development [11]. In the initial identification of the S1P1 receptor,

induction of edg1 transcripts occurred in differentiating human endothelial cells,

suggesting a role for this receptor in vascular development [71]. Also, the S1p1-knockout

mouse is embryonic lethal with the observed hemorrhage of vessels resulting from the

failure of the vascular smooth muscle to form dorsally [84]. In agreement with the S1p1-

knockout mouse phenotype, activation of the S1P1 receptor induces cell-cell contact

through N-cadherin (component of adherens junctions) recruitment in endothelial and

mural cells to the plasma membrane, promoting vascular stabilization during vessel

formation [85]. Also, vascular endothelial cells in culture with a stable knockdown of

S1P1, were no longer capable of forming capillary structures [86].

Unlike the S1p1-knockout mouse that is embryonic lethal between E12.5 and

E14.5, the S1p3-knockout mouse shows no obvious phenotype [87] and about 20% of the

S1p2-knockouts experienced an epileptic seizure [88], while deafness was detected in

others [89]. About 50% of the animals lacking both the S1P2 and S1P3 receptors die after

Page 33: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

32 

 

E13.5; animals lacking S1P1 and S1P2 die between E10.5 and E12.5 while animals

without S1P1-3 die between E10.5 and E11.5 [87, 90]. All S1P receptor knockout animals

with embryonic lethality and double null animals that survived displayed vascular

defects, demonstrating the S1P1-3 receptors to be key players in angiogenesis and vascular

maturation. The viable S1p2 and S1p3-knockout animals, along with the differences in

timing of embryonic lethality between the knockout animals, suggest redundant and

compensatory signaling among the S1P receptors. As described below, the importance of

the S1P receptors in angiogenesis and vascular maturation is due to the regulation of

cytoskeletal dynamics and cell contacts by downstream effectors of S1P signaling.

2.4.3 Sphingosine-1-phosphate effects on the cytoskeleton and junction assembly

S1P receptor signaling plays a major role in regulating cytoskeletal dynamics

[13]. The key proteins downstream of S1P receptor activation that regulate cytoskeletal

dynamics are members of the Rho GTPase family, Rac and Rho [91]. The Rho GTPases,

a family composed of about 20 members (RhoA, Rac1, and Cdc42 being the most

studied), are part of the Ras superfamily of proteins that are characterized by their

intrinsic guanosine triphosphatase activity, highlighted by their switching between GDP

and GTP-binding motifs [92]. The Rho-GTPases play a major role in cell migration by

regulating actin cytoskeleton rearrangement [93]; Rac promotes lamellipodia formation at

the leading edge, Cdc42 promotes filopodia formation to ‘sense’ the cell surroundings,

and Rho promotes stress fiber formation and cellular contraction to move the cell body.

S1P promotion of cell migration occurs through the S1P1 and S1P3 receptors [94-97]

Page 34: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

33 

 

while the S1P2 receptor inhibits migration [98-100]. In particular, S1P1 activates Rac,

promoting lamellipodia formation and initiating cell migration [84, 101]. On the other

hand, S1P2 activates Rho and inhibits Rac, thereby preventing migration [98-100]. The

S1P3 receptor seems to activate both Rac and Rho, but unlike S1P2, does not prevent

migration by inhibiting Rac [102]. Thus, S1P-induced cell migration requires a balance

between Rac and Rho activation to promote cytoskeletal rearrangement in cell spreading

and cell-matrix adhesion/contraction.

In addition to cell migration, the Rac and Rho GTPases play a major role in the

cytoskeletal rearrangement that helps regulate endothelial permeability [103]; Rho

generally promotes actomyosin contraction and increases permeability by disrupting cell-

cell junctions, while Rac decreases permeability by promoting cortical actomyosin

formation and stabilizing the junctions. S1P has been shown to increase vascular barrier

integrity by activating Rac and increasing junctional organization [13, 104]. Specifically,

S1P promotes cortical actin rearrangement with an associated increase in myosin light

chain (MLC) phosphorylation, and cortactin and MLC kinase (MLCK) recruitment to the

cell cortex [17, 19, 105]. The increased actomyosin organization at the cell cortex

provides structural support while a concurrent increase in junctional proteins such as

vascular endothelial cadherin, β-catenin, and zonula occludens-1 occurs at sites of cell-

cell contact [14-16]. Along with stabilizing the cell-cell contacts, S1P-induced

cytoskeletal rearrangement also helps stabilize the cell-matrix junctions [18, 106]. The

Rac-dependent cortical actin formation is due to S1P activating the S1P1 receptor [107].

The subsequent increase in endothelial barrier formation though, seems to involve both

Page 35: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

34 

 

the S1P1 and S1P3 receptors activating both the Rac and Rho pathways [15, 17]. While

Rac promotes cortical actin formation and recruitment of junction proteins to sites of cell-

cell contact, Rho promotes strengthening of the actomyosin cytoskeletal network through

the Rho/Rho-kinase pathway that inhibits MLC phosphatase activity. MLC exists in an

inactive and active state where phosphorylated MLC drives myosin and actin cross-

bridge cycling. The state of MLC is determined by the activity of the MLC phosphatase

that de-phosphorylates MLC, and the MLCK that phosphorylates MLC through a calcium

and calmodulin-dependent pathway [108, 109]. Thus, activation of the Rho/Rho-kinase

pathway enhances MLC phosphorylation and increases the contractile state of the cell

(Figure 2.3).

Figure 2.3. MLC phosphorylation and cell contractility through RhoA activation. GTP-bound RhoA activates Rho-kinase, inducing the phosphorylation and inactivation of MLC phosphatase. With MLC phosphatase activity blocked, phospho-MLC species (MLC-P) are increased and promotes cell contractility. Phosphorylation of MLC occurs through the calcium (Ca2++)/calmodulin-dependent MLC kinase, a process requiring an increase in intracellular calcium.

The initial driving force increasing MLC phosphorylation by MLCK is an

increase in intracellular calcium. Increased intracellular calcium in endothelial cells

occurs through receptor-mediated activation of the Gq protein, which promotes

Page 36: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

35 

 

phospholipase C to cleave phosphatidylinositol 4,5-bisphosphate into diacylglycerol

(DAG) and inositol triphosphate (IP3). IP3 binds to IP3 receptors on the endoplasmic

reticulum to release calcium from intracellular stores while DAG activates the TRPC6

channel to increase calcium influx from the extracellular space [110]. Interestingly, both

the S1P2 and S1P3 receptors couple to the Gq protein [82, 83]; thus, S1P receptor

activation may enhance MLC phosphorylation by inactivating the MLC phosphatase

through the Rho/Rho-kinase pathway and enhancing MLCK activity through an increase

in intracellular calcium.

While S1P1 and S1P3 receptor binding promotes endothelial barrier enhancement

through Rac and Rho activation [13], S1P2 decreases barrier integrity. As mentioned

previously, S1P2 activates the Rho pathway while inhibiting the Rac pathway, thereby

preventing cell migration [98-100]. The activation of the S1P2 receptor promotes a Rho-

dependent MLC phosphorylation with an increase in stress fiber formation and cell

contractility [111-113]. In particular, S1P2 has been shown to increase endothelial

permeability by disrupting vascular endothelial-cadherin junctions, a process prevented

by the S1P2-specific antagonist JTE-013 [114]. Interestingly, hypoxic conditions in the

mouse retina seems to promote vascular permeability and activate an inflammatory

pathway through the S1P2 receptor, a process triggering angiogenesis [115].

2.4.4 Sphingosine-1-phosphate receptor compounds

The S1P system is involved in many biological processes and various compounds

specific for the S1P receptors have been synthesized and are being used not only as

Page 37: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

36 

 

research tools, but also as therapeutic options [116]. In the immune system, S1P1

receptor activation is required for lymphocytes to exit and re-circulate to the various

secondary lymphoid tissues. The S1P1,3,4,5 agonist FTY720 (fingolimod) has been shown

to be an effective drug in inhibiting lymphocyte egress to suppress the immune response

[117, 118]. The mechanism occurs through binding of the phospho-FTY720 (FTY720 is

phosphorylated by SK in vivo) to the S1P1 receptor and down-regulating its expression on

the lymphocyte cell surface, thus reducing S1P1 receptor signaling and sequestering the

lymphocytes in lymph nodes. FTY720 has emerged in human clinical trials as an

effective therapeutic for relapsing multiple sclerosis by suppressing lymphocyte

circulation [119]. Additionally, the S1P1-specific agonist SEW2871 has been shown to

protect renal function by also suppressing lymphocyte circulation and decreasing pro-

inflammatory molecules in the mouse renal ischemia/reperfusion (I/R) model [120].

In addition to S1P receptor agonists, commercial antagonists have also been

developed. For example, the S1P1 antagonist W146 decreased capillary leakage in both

mouse skin and lung, demonstrating the importance of S1P1 in maintaining barrier

integrity [121]. JTE-013 was shown to bind the S1P2 receptor with high affinity [122]

and prevent S1P2- mediated vascular permeability [114]. Some of the commercially-

available S1P receptor compounds currently available are shown in Figure 2.4.

Page 38: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

37 

 

Figure 2.4. Chemical structures of subtype-specific S1P receptor compounds. Commercially available S1P receptor agonists include FTY720 (S1P1,3,4,5-specific) and SEW2871 (S1P1-specific). S1P receptor antagonists include W146 (S1P1-specific), JTE-013 (S1P2-specific), and VPC23019 (S1P1,3-specific). Figures of S1P, FTY720, SEW2871, JTE-013, and VPC23019 were previously published in Biochemical Pharmacology: Huwiler, A. and Pfeilschifter, J. (2008). New players on the center stage: Sphingosine-1-phosphate and its receptors as drug targets. Biochem Pharmacol. 75:1893-1900. Used with permission from Elsevier. Figure of W146 was previously published in Immunological Reviews: Rosen, H. et al. (2008). Modulating tone: the overture of S1P receptor immunotherapeutics. Immunol Rev. 223: 221-35. Used with permission from John Wiley and Sons.

Page 39: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

38 

 

2.5 Sphingosine-1-phosphate receptor signaling and the conventional outflow pathway

S1P receptors have been identified at the transcript level in both human TM cell

culture and tissue-derived libraries [9]. In the same study, S1P was shown to decrease

outflow facility in the porcine whole-eye perfusion model while in TM cells, promote

Rho activation and increased cell contractility through MLC phosphorylation, focal

adhesion, and stress fiber formation [9]. These results suggest that a S1P-promoted

contraction of the TM, with subsequent rearrangement of aqueous humor flow patterns, is

responsible for S1P increasing outflow resistance.

Although TM cells have been primarily investigated, the effects of S1P on SC

cells in culture and whether similar or different downstream effectors are activated have

not. Due to the inner wall of SC being a continuous endothelial monolayer with vascular

developmental origins [123], S1P may promote a similar decrease in endothelial

permeability, thus increasing resistance. Promotion of endothelial barrier and junctional

formation along the inner wall may increase trans-endothelial pressure across the

monolayer and result in the formation of giant vacuoles. Indeed, an increase in giant

vacuole formations across the inner wall of the aqueous plexus (analogous to inner wall

of SC) was observed in S1P-treated porcine eyes compared to control, supporting the

inner wall as a key contributor in the S1P-induced increase in outflow resistance.

Page 40: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

39 

 

3. SPHINGOSINE-1-PHOSPHATE EFFECTS ON THE INNER WALL

OF SCHLEMM’S CANAL AND OUTFLOW FACILITY IN PERFUSED

HUMAN EYES

This chapter has been published in Experimental Eye Research: Stamer, W.D.,

Read, A.T., Sumida, G.M., and Ethier CR. (2009). Sphingosine-1-phosphate effects on

the inner wall of Schlemm’s canal and outflow facility in perfused human eyes. Exp Eye

Res, 89: 980-988. PMCID: PMC2794662. Used with permission from Elsevier.

3.1 Introduction

Lysophospholipids, such as sphingosine-1-phosphate (S1P), are membrane

phospholipid metabolites that can function as autocrine/paracrine signaling molecules,

influencing a broad range of cellular functions, such as cardiac development, immunity,

platelet aggregation, cell movement and vascular permeability [116, 124]. S1P activity is

mediated by binding to one or more of five G-protein coupled receptor subtypes (S1P

receptors 1–5, formerly known as Edg1, 3, 5, 6, 8). The S1P receptor subtypes are

differentially expressed in tissues, likely in alignment with specific functional tissue

requirements. For example, S1P1 and S1P3 receptors are preferentially expressed by

vascular endothelial cells, whereas smooth muscle cells express S1P1, S1P2 and S1P3

[13]. Due to the fact that it is constantly bathed by secreted aqueous humor, the

conventional outflow pathway has the potential to utilize S1P and/or other

Page 41: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

40 

 

lysophospholipids as signaling molecules to modulate outflow resistance. In support of

this idea, lysophospholipids are known to be constituents of aqueous humor [125] and it

has been shown that activation of S1P receptors in the conventional outflow tract

dramatically and rapidly decreases outflow facility in porcine eyes. Specifically, outflow

facility decreased by 31% in perfused porcine eyes after 5 hours of infusion of 5µM S1P

[9]. Interestingly, while Mettu et al. observed no histological changes in the

juxtacanalicular region of the TM of perfused porcine eyes, they did observe a dramatic

increase in the density of giant vacuoles in the endothelial lining of the angular aqueous

plexus (the porcine analogue of SC in human eyes). This observation is consistent with

S1P affecting the pressure drop across the endothelial lining, perhaps by increasing the

strength of cell-cell junctions between the endothelial cells lining the aqueous plexus.

This premise is in turn consistent with well-described effects of S1P on cell-cell junction

and circumferential actin assembly in endothelial cells via downstream effects on the

small GTPase, Rac1 and subsequently decreased paracellular permeability [17-19].

Despite obvious effects on cells lining the aqueous plexus, S1P receptor expression and

activation has only been studied in TM cells in culture, where it was shown that TM cells

express S1P1 and S1P3 receptor subtypes [9]. Further, Mettu et al. showed that S1P

changed several measures of TM contractility, e.g. S1P promoted the phosphorylation of

myosin light chain plus the formation of stress fibers and focal adhesions, which were

shown to be mediated primarily through Rho GTPase activation. Due to apparent rho-

dominant signaling in TM cells, it was concluded that S1P3 receptors mediate S1P effects

on TM cell contractility and hence likely affect outflow facility in the porcine eye.

Page 42: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

41 

 

Motivated by this important work in porcine eyes, we sought to determine

whether S1P affects outflow facility in human eyes, and if so, what role S1P receptor

subtypes in the inner wall of SC might play in this process. We hypothesized that S1P

increases outflow resistance in human eyes by activating receptors in the inner wall of

SC, driving circumferential actin and associated cell-cell junction assembly. In the

present study, we observed that, similar to porcine eyes, S1P dramatically and rapidly

decreases outflow facility in enucleated human eyes. However, unlike the situation in

porcine eyes, the inner wall of SC in treated human eyes was not morphologically

different from untreated eyes. At the molecular level, the inner wall of SC expressed

S1P1 and S1P3 receptor subtypes, but activation of these receptors did not result in

detectible changes in cortical actin, VE-cadherin, phosphotyrosine or β-catenin

distribution/abundance.

Page 43: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

42 

 

3.2 Methods

3.2.1 Cell culture

Human donor eyes were obtained from Life Legacy Foundation (Tucson, AZ),

National Disease Research Interchange (Philadelphia, PA) and Sun Health Research

Institute (Sun City, AZ). SC cells were isolated from conventional outflow tissues of

human eyes using a cannulation technique and then were characterized and cultured as

previously described [126]. Using a blunt dissection procedure followed by extracellular

matrix digestion, TM cells were isolated from human eyes and were characterized and

cultured as previously described [127]. Cells were maintained in Dulbecco’s modified

Eagle’s medium (DMEM, low glucose), supplemented with 10% fetal bovine serum,

penicillin (100 units/ml), streptomycin (0.1 mg/ml) and glutamine (0.29 mg/ml). Six

different SC cell strains (SC42, SC44, SC45, SC51, SC55, and SC 56) and four TM cell

strains (TM26, TM86, TM87, and TM90) were used in the present study and chosen

based on strain availability at the time of experiments.

3.2.2 Reverse-transcription polymerase chain reaction

Total RNA was extracted from cell strains using the TRIzol reagent (GIBCO).

RNAs were used as templates for reverse transcription synthesis of cDNAs using the

ThermoScript RT-PCR kit (Invitrogen). Amplification of DNA by the polymerase chain

reaction (PCR) was performed using Taq DNA polymerase (Invitrogen) for 30 cycles

(denaturation at 94˚C for 30s, annealing at 55˚C for 30s, and extension at 72˚C for 45s).

Oligonucleotide primers used for subtype-specific amplification to detect the S1P

Page 44: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

43 

 

receptors were previously characterized [9]: edg1 (5’-

ACGTCAACTATGATATCATCGTCCG, 3'-CATTTTCAGCATTGTGATATAGCGC);

edg5 (5'-ACTGTCCTGCCTCTCTACGCC, 3'-GTCTTGAGCAGGGCTAGCGTC); and

edg3 (5'-ACCATCGTGATCCTCTACGCAC, 3'-

CTTGATTTACTTCTGCTTGGGTCG). Positive control primers were directed against

gapdh (GAPDH6- GAAGGTGAAGGTCGGAGTC, 3’GAPDH 212-

GAAGATGGTGATGGGATTTC). PCR products were loaded into 1% agarose gel

slabs, resolved by electrophoresis and visualized using ethidium bromide and ultraviolet

light.

3.2.3 Western blot analyses

Mature and confluent SC and TM cell monolayers were scraped from culture

plates and solubilized in Laemmli sample buffer containing 10% β-mercaptoethanol. The

whole cell lysates were boiled for 10 minutes, loaded onto a 10% polyacrylamide gel, and

proteins were fractionated by SDS-PAGE. Proteins were electrophoretically transferred

from gel slabs to nitrocellulose membranes for 90 min at 100 V. Membranes were then

blocked with 5% non-fat dry milk in Tris-buffered saline (137 mM NaCl, 25 mM Tris,

2.7 mM KCl, pH 7.4) containing 2% Tween-20 (TBS-T) for 60 min, then incubated with

rabbit polyclonal IgGs that specifically recognize S1P1 (0.1 μg/ml, Affinity Bioreagents),

S1P2 (EDG5, 0.2 μg/ml, Santa Cruz), or mouse monoclonal IgGs against S1P3 (EDG3,

0.1 μg/ml, Exalpha Biologicals) receptor subtypes. Following overnight incubation at

4˚C, membranes were washed (3 x 10 min) with TBS-T, incubated with horseradish

Page 45: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

44 

 

peroxidase (HRP)-conjugated goat anti-rabbit or goat anti-mouse IgGs (40 ng/ml,

Jackson Immunoresearch Laboratories) for 60 min at room temperature, and washed

again with TBS-T (3 x 10 min). To visualize proteins, membranes were incubated with

either ECL Advance (Amersham) or HyGLO (Denville Scientific) chemiluminescence

reagents and exposed to X-ray film (Genesee Scientific). All membranes were re-probed

with ascites fluid containing a mouse monoclonal IgG against β-actin (1:10,000 dilution,

Sigma-Aldrich) and subsequent HRP-conjugated goat anti-mouse IgG (40 ng/ml, Jackson

Immunoresearch Laboratories) for loading control. S1P1-specific bands were determined

with the inclusion of EDG1 transfected cell lysates (Exalpha Biologicals).

3.2.4 Immunofluorescence microscopy of frozen ocular sections

Human cadaveric eyes (donor ages= 79, 88 and 98) were received within 36 hours

of death and their anterior portions were dissected into radially oriented wedges. Tissue

wedges were immersed in OCT compound, frozen at -80ºC and then saggitally

cryosectioned (8 µm). The tissue sections on slides were fixed in 4% paraformaldehyde,

then blocked for 30 min with 10% goat serum in 100 mM Tris-HCl containing 0.05%

Tween-20, pH 7.4. The sections were then incubated overnight in a moist chamber at

4˚C with rabbit polyclonal IgGs that recognize S1P1 (EDG1, 4 μg/ml, Santa Cruz), S1P2

(EDG5, 4 μg/ml, Santa Cruz), or S1P3 (EDG3, 4 μg/ml, Santa Cruz) receptor subtypes.

Following antibody incubations, sections were washed extensively with 100 mM Tris-

HCl containing 0.05% Tween-20 (4 x 10 min). Antigen binding was detected by a 1 hr

incubation with CY3-conjugated goat anti-rabbit IgG (0.75 μg/ml, Jackson

Page 46: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

45 

 

Immunoresearch Laboratories), counterstained with SYTOX green nucleic acid stain

(100 nM, Invitrogen) for 1 minute, and washed extensively (4 x 10 min) before

visualization. Background and auto- fluorescence was monitored by incubating tissues

with CY3-conjugated goat anti-rabbit IgG in the absence of primary antibodies. Labeled

tissue sections were visualized and captured digitally using a Nikon PCM 2000 confocal

microscope (Melville, NY).

3.2.5 Whole-eye perfusion and facility measurements

Dulbecco's phosphate buffered saline containing 5.5 mM glucose (DBG) and pre-

filtered through a 0.22 µm Millex-GS filter (Millipore, Bedford, MA) served as mock

aqueous humor for the perfusion studies. S1P, acquired from Biomol International

(Plymouth Meeting, PA), was dissolved in 65ºC boiling methanol (0.5mg/mL) and

aliquotted into glass tubes. The solvent was evaporated with streaming nitrogen gas to

deposit a thin film inside the tube and the aliquots stored at -20ºC. Working solutions of

5µM S1P were obtained by dissolving the stock aliquot into DBG containing 0.2% fatty-

acid free BSA (Roche, Penzberg, Germany) on a rotator overnight at 37ºC. Control

solutions also contained 0.2% fatty-acid free BSA.

Ostensibly normal human eyes were obtained from the Eye Bank of Canada

(Ontario Division, Toronto, Ontario) and NDRI (Philadelphia, PA). After excluding six

pairs of eyes with unstable or asymmetric baseline facility traces, 10 pairs remained in

the study. Mean donor age was 79.8 years (range 74-92 years) and mean post mortem

Page 47: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

 

 

46

Table 3.1. Summary of Eye Donor Characteristics and Perfusion Results with S1P

ID Donor Age (yrs)

Donor Gender

Time to Enucleation (hrs)

Time to Perfusion (hrs)

Baseline C (μl/min/mmHg)

Ending C (μl/min/mmHg)

Increase in C (%)

Net Change in C (%)

797-e 79 F 0.5 20 0.21 0.1 -52.4 -32.4 798-v 0.15 0.12 -20.0 811-e 75 M 2.3 35.8 0.2 0.14 -30.0 -87.9 812-v 0.19 0.3 57.9 813-e 74 F 2.3 33 0.3 0.19 -36.7 -40.2 814-v 0.29 0.3 3.5 683-e 92 F 3.2 27 0.23 0.18 -21.7 -21.7 684-v 0.19 0.19 0 695-e 75 F 7.2 38.3 0.3 0.13 -56.7 -36.7 696-v 0.15 0.12 -20.0 697-v 83 M 2.0 28.5 0.22 0.19 -13.6 -40.6 698-e 0.24 0.11 -54.2

6101-v 76 M 4.0 32 0.28 0.25 -10.7 -16.6 6102-e 0.33 0.24 -27.3 6105-e 78 F 1.5 38.5 0.26 0.22 -15.4 -23.3 6106-v 0.38 0.41 7.9 7025-v 76 M NA 39 0.26 0.24 -7.7 -42.3 7026-e 0.24 0.12 -50.0 8005-e 90 F NA 15.5 0.26 0.14 -46.2 -21.2 8006-v 0.24 0.18 -25.0

79.8 6F/4M 2.9 30.8

Control mean 0.24 0.23 -2.8 -36.3

S1P-treated

mean 0.26 0.16 -39.1

Page 48: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

47 

 

time to start of perfusion was 30.8 hours (range 15.5-39.0 hours) (table 1). DBG was

introduced into eyes at a constant pressure of 8 mmHg (corresponding to 15 mmHg in

vivo) via a needle inserted through the cornea to the posterior chamber as described

previously [128]. Baseline facility was measured for 60-90 min and then one eye of each

pair received anterior chamber exchange with 5 µM S1P, while the contralateral eye was

exchanged with DBG containing only vehicle. During the anterior chamber exchange a

constant IOP of 8 mmHg was maintained after which the perfusion was allowed to

continue for approximately 180 min at 8 mmHg. The percent increase in outflow facility

(C) is 100*(C final/C Baseline – 1) and net change in C is percent increase in experimental

eye minus percent increase in control eye. Average net change values were analyzed by a

two-tailed, paired Student’s t-test assuming unequal variance and differences were

considered significant at p<0.05.

Following S1P perfusion, eyes were fixed by anterior chamber exchange and

perfusion with 3% formalin at 8 mmHg, then hemisected, and the outflow tissue cut into

wedges. Half from each eye, including tissue wedges from every quadrant, were

immersed overnight in universal fixative (UF, 2.5% formalin, 2.5% glutaraldehyde in

Sorensen’s buffer) for use in scanning electron microscopy (SEM) and semi-thin

sectioning. The rest, to be used for confocal microscopy, were fixed in 3% formalin, the

exception being eyes 07-025/026 and 08-005-006, which were perfusion fixed only, then

stored in 15% glycerol in DBG at -20ºC.

Page 49: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

48 

 

3.2.6 Labeling and confocal microscopy of the inner wall of SC

Radial segments of the limbal area were microdissected and labeled as previously

described [129]. Briefly, SC was opened by an incision along its posterior margin, and

the TM and adherent inner wall reflected anteriorly. The inner wall and underlying

trabecular meshwork were fluorescently labeled to visualize F-actin, nuclei, and either

VE-cadherin, β-catenin or phosphotyrosine. Tissue was permeabilized with 0.2% Triton

X-100 in DPBS for 5 min at room temperature (RT) and blocked with 5% goat serum

(Sigma Corp., St. Louis, MO) in DPBS for 45 min at 37ºC. VE-cadherin was labeled with

mouse anti-VE-cadherin IgG (clone 9H7, provided by Dr. Ron Heimark) diluted in

calcium- and magnesium-free DPBS, and incubated overnight at RT, followed by

incubation in Alexa-647 goat anti-mouse IgG (Invitrogen, Austin, TX) diluted 1:150 in

DPBS, for 75 min at 37ºC. β-catenin was labeled with rabbit anti-β-catenin IgG (Abcam,

Cambridge, MA), diluted 1:200 in DPBS, and incubated for 2h at RT, followed by

incubation in Alexa-647 goat anti-rabbit IgG (Invitrogen, Austin, TX) diluted 1:150 for

75 minutes at 37ºC. Phosphotyrosine was labeled with mouse anti-phosphotyrosine IgG

(clone 4G10; Upstate, Lake placid, NY), diluted 1:200 and incubated at 37ºC for 60 min

followed by incubation in Alexa-647 goat anti-mouse IgG (Invitrogen, Austin, TX). For

negative controls, tissue was treated as above while excluding the primary antibodies. F-

actin was labeled by incubating for 30 min at RT in rhodamine-phalloidin (Invitrogen,

Austin, TX), diluted 1:40 in DPBS. Nuclei were marked by incubating for 5 min at RT in

SYTOX (Invitrogen, Austin, TX), diluted 1:2400 in Tris-buffered saline or DAPI

(Invitrogen, Austin, TX) diluted 2 µg/mL in DPBS.

Page 50: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

49 

 

The thin, 2 mm circumferential section of tissue comprising the inner wall of SC,

JCT and TM was separated from the ciliary body and root of the iris by an oblique cut at

the posterior margin of SC, then mounted with DAKO® fluorescent mounting medium

(DAKO, Glostrup, Denmark) and a No. “0” cover slip. Samples were examined using a

Zeiss LSM 510 meta confocal microscope (Carl Zeiss Inc., Jena, Germany). Six pairs of

perfused eyes (7025/6, 8005/6,683/4, 695/6, 697/8, 6109/10) were examined in a masked

fashion by three observers, and the average number of preparations examined was eight

per eye. Z-series were collected using a x63 oil or water immersion lens, with the focal

plane beginning at the apex and advancing deeper into the tissue. Images were viewed

using Zeiss Image Browser software, version 5.

3.2.7 SEM and semi-thin sectioning

Outflow tissue from eyes that had been fixed in UF was dissected out, post-fixed

in 1% osmium tetroxide (EMS, Hatfield, PA), dehydrated, infiltrated, and embedded in

Epon-Araldite (EMS, Hatfield, PA). Half-micron thick radial sections were stained with

toluidine blue, examined and photographed with a Zeiss Axiovert microscope. Samples

from all four quadrants of four pairs of eyes were examined in detail (6105/6, 683/4,

813/4, 697/8).

For SEM, inner wall samples from all four quadrants in two pairs of eyes were

examined in detail (6105/6 and 813/4). Eyes were micro-dissected to expose the inner

wall of SC, as described above, fixed in UF, incubated in a 2% solution of guanidine-

HCl/tannic acid (Sigma Corp., St. Louis, MO) for 2h, post-fixed in 1% osmium tetroxide

Page 51: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

50 

 

for 1h, washed then dehydrated though an ethanol series, incubated in

hexamethyldisilizane (Sigma Corp., St. Louis, MO) and air dried in a fume hood. They

were mounted on SEM stubs, gold-coated and examined with a Hitachi S-3400N VP

SEM (Pleasanton, CA).

Page 52: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

51 

 

3.3 Results

We used three complementary approaches to test the hypothesis that S1P effects

on conventional outflow in human are mediated at the level of cell-cell junctions between

cells of the inner wall of SC. Using primary cultures of human SC endothelial cells, we

first examined the expression of three S1P receptor subtypes (S1P1-3) via RT-PCR and

Western blot analyses. Similar to porcine TM cells [9], we observed that SC cells

express messenger RNA for all three receptor subtypes tested (Table 3.1), but only

protein for S1P1 and S1P3 receptors (Figure 3.1).

These results are consistent with data gathered from confocal indirect

immunofluorescence studies using subtype-specific antibodies to label S1P receptors in

human cadaveric eyes (Figure 3.2). We observed the presence of S1P1 and S1P3 receptor

subtypes in the SC endothelia. These studies also confirmed that both S1P1 and S1P3

receptor subtypes were expressed by cells on the trabecular beams and in the

juxtacanalicular tissue. Interestingly, labeling of S1P1 and S1P3 receptors appeared

stronger on SC endothelia than on TM cells in all donor eyes examined. In two of three

eyes analyzed, we did notice some non-specific labeling of S1P3 antibodies to non-

cellular material of sclera. Additionally, some modest SC and TM labeling cells was

observed using antibodies that recognize S1P2 receptors (Figure 3.2).

Page 53: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

52 

 

Table 3.2 Summary of RT-PCR results

SC TM Receptor    SC42 SC44 SC45 SC51    TM86 TM87 TM90

S1p1 + + + + + + +

S1p2 + + + - + + -

S1p3    + + + - + + +

(+) indicates positive expression while (-) indicates negative expression

Figure 3.1. Western blot analyses of S1P receptor subtype expression in monolayers of Schlemm’s canal (SC) endothelial cells. Whole cell lysates derived from three mature SC cell monolayers (strains SC44, SC55, and SC56) were probed with antibodies specific for S1P1, S1P2 and S1P3 receptor subtypes. Whole cell lysates prepared from human microvascular endothelia (HM), human umbilical vein endothelia (HV) and human trabecular meshwork (TM26) cells were also analyzed and used as controls. Blots were re-probed with antibodies specific for β-actin to control for equivalent loading between samples.

Page 54: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

53 

 

Figure 3.2. Expression of S1P receptor subtypes by human Schlemm’s canal (SC) endothelia in situ. Confocal immunofluorescence microscopy of frozen serial saggital sections of human anterior segments shows labeling with antibodies specific for S1P1, S1P2 and S1P3 receptor subtypes (as indicated, red) and cell nuclei (SYTOX, green). Insets indicate internal positive control staining for S1P1 and S1P3 receptors located on scleral vessels (V) in same tissue section. The lower right panel is provided to show background staining of tissues labeled only with rhodamine-conjugated secondary antibodies and nuclei stain (control). Arrows point to labeling in trabecular meshwork (TM). Results shown are all from one human donor eye and are representative of three different human eyes analyzed.

Page 55: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

54 

 

Figure 3.3. Effect of S1P on outflow facility in paired whole human eye perfusions. After one hour of relatively stable facility, anterior chamber contents in one eye were exchanged with medium (DPBS + 5.5 mM glucose + 0.2% fatty-acid free BSA) containing 5 µM S1P while the contralateral eye was exchanged with medium only, and then perfused for an additional 3.5 hours (post-exchange). The plotted quantity is measured outflow facility normalized by each eye’s stable facility value just prior to exchange (mean ± SEM for ten pairs of eyes). Normalized facility values less than one therefore indicate a decrease in facility compared to baseline.

Next, we examined whether S1P affects outflow facility in human eyes in a

fashion similar to that reported for porcine eyes. We tested 10 pairs of enucleated human

donor eyes (Table 3.2) in which stable baseline outflow facilities were found to be similar

between control (0.235±0.07 µl/min/mmHg) and their experimental counterparts

(0.257±0.04 µl/min/mmHg) and within a physiological range (Table 3.2). Our results

showed that S1P significantly decreased outflow facility by 36 ± 20% (Figure 3.3;

Page 56: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

55 

 

p=0.0004), which was similar in magnitude to that reported for porcine eyes.

Impressively, S1P effects were observed very rapidly, within 20 minutes of anterior

chamber exchange.

Figure 3.4. Morphological analyses of the inner wall of Schlemm’s canal and associated conventional outflow structures in human eyes perfused with S1P. Shown are semi-thin (plastic) sections of outflow tissues from a control (panel A) and S1P-perfused (panel B) eye from a pair, fixed at 8 mmHg, saggitally sectioned (0.5 µm) and stained with toluidine blue (bar= 100 µm). Micrographs are from a representative eye pair (6105/6) of four that were examined in detail. The inner wall of SC (viewed from the luminal side) in perfused eyes was examined by scanning electron microscopy. Displayed in panel C is a micrograph taken from a control eye, while panel D was taken from the contralateral, S1P-treated eye (6105/6). Results shown are representative of 2 eye pairs that were examined by SEM in all four quadrants (bar=25 µm). Representative giant vacuoles are indicated by arrowheads.

Page 57: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

56 

 

Figure 3.5. Actin architecture and Vascular Endothelial (VE)-cadherin expression in Schlemm’s canal inner wall of eyes perfused with S1P. Post perfusion, outflow tissues were probed with rhodamine phalloidin to label filamentous actin (red) and monoclonal antibodies specific for VE-cadherin (green). Shown are confocal images taken of a single area of human SC inner (viewed from luminal side) wall in paired eyes. Merged images are shown in right panels. Shown are micrographs from a single eye pair (7025/026) to indicate variability that was observed in staining patterns between S1P-treated and control eyes. Data shown are representative of six pairs of eyes that were examined.

Page 58: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

57 

 

Finally, we studied whether S1P treatment induced morphological changes in

outflow tissues (including SC) of perfused human eyes, and whether such changes were

similar to those reported in porcine eyes [9]. We first examined saggital sections in each

of four quadrants of perfused conventional outflow tissues by light microscopy. In all

cases, no significant differences in inner wall morphology between S1P-treated eyes and

their paired controls were observed (Figure 3.4). Overall, outflow structures were

populated by appropriate numbers of cells for the age of the donors, the cells appeared

healthy and the inner wall was intact in both control and treated eyes. Moreover, giant

vacuole density was examined qualitatively and found not to be different between treated

and control eyes. We also studied the morphology of the inner wall en face using

scanning electron microscopy. Although we did not quantify giant vacuole or pore

density in the inner wall, no notable differences between S1P-perfused and control eyes

were observed, with each displaying similar vacuole densities, vacuole sizes/shapes,

inner wall pore appearances and cell-cell junction appearances (Figure 3.4).

To explore whether the decreased outflow facility in S1P-treated eyes could be

linked with changes at the molecular level, we used indirect immunofluorescence

confocal microscopy to study the expression and cellular distribution of several

components known to influence endothelial permeability: F-actin, VE-cadherin, β-catenin

and phosphotyrosine. While we did notice circumferential reorganization of F-actin in

some S1P-treated eyes compared to their contralateral controls, these observations were

not consistent amongst all treated eyes (Figures 3.5 and 3.6). Similarly, VE-cadherin

labeling (Figure 3.5), β-catenin labeling (Figure 3.6) and phosphotyrosine labeling (data

Page 59: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

58 

 

not shown) appeared unaltered by S1P treatment. In fact, extensive masked analysis of

labeled tissues failed to identify any constant effect of S1P on the distribution or amount

of the above factors.

Figure 3.6. Actin architecture and β-catenin expression in Schlemm’s canal inner wall of eyes perfused with S1P. Post perfusion, outflow tissues were probed with rhodamine phalloidin to label filamentous actin (red) and monoclonal antibodies specific for β-catenin (green). Shown are confocal images taken of a single area of human SC inner (viewed from luminal side) wall in paired eyes perfused with S1P or vehicle control. Merged images are shown in right panels. Shown are micrographs from a representative eye pair (8005/6) of two pairs of eyes that were examined in total.

Page 60: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

59 

 

3.4 Discussion

This study demonstrates that S1P causes rapid and substantial decreases in

outflow facility in human eyes. Because receptors for S1P were found on both TM and

SC cells in the conventional outflow pathway, the involvement of both cell types in the

S1P response is possible. Interestingly, even though S1P receptors appeared more

abundant in SC endothelia, we did not find morphologic alterations of inner wall cells in

human eyes similar to the dramatic ballooning of aqueous plexus endothelial cells

observed in porcine eyes treated with the same dose and time course of S1P [9]. In fact,

we could observe no consistent morphological or molecular changes to explain the

facility decrease due to S1P perfusion in human eyes. This suggests that a number of

subtle synergistic changes in outflow tissues are responsible for S1P’s rapid and

substantial effect on facility.

While the time course and magnitude of facility changes in response to S1P were

remarkably similar between porcine [9] and human eyes, the reason(s) for the differences

in giant vacuole density between these two species is unclear. Differences in physiology

between the porcine and human outflow tracts (e.g. greater connectivity between the

JCT-TM cells and inner wall of SC is observed in human eyes compared to other species

[130, 131]) may underlie dissimilarities. Alternatively, there may be a species difference

in the distribution and/or abundance of S1P receptors between TM and SC cells.

A limitation of this study was that we did not quantify giant vacuole density or

expression levels of VE-Cadherin and β-catenin. This was primarily due to technical

issues; for example, quantification of giant vacuoles from SEM is problematic due to

Page 61: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

60 

 

issues with differentiating between nuclear bulges and giant vacuoles, necessitating time-

consuming analysis of serial micrographs. Moreover, for analyses of VE-cadherin and β-

catenin, the small volume of TM/SC tissues requires tissue pooling for quantitative

analysis of protein levels. Thus, we cannot exclude the possibility that there were subtle

changes in these endpoints that were not evident by qualitative inspection, although

considering the degree of heterogeneity around the circumference of the TM; it is our

experience that changes typically need to be qualitatively evident to be statistically

robust.

Activation of S1P receptors in a contractile cell increases cellular tone; S1P would

therefore be expected to increase TM cell tone, which is predicted to decrease outflow

facility [132, 133]. Further, S1P increases barrier function in endothelia, and therefore

would be expected to decrease the permeability of SC cells. Since activation of S1P

receptors in both cell types is predicted to decrease outflow facility, it is possible that

both TM cells and SC cells participate in the S1P-mediated effects observed in human

whole eye perfusions. At this time the relative contribution of each cell type to the total

facility change is uncertain.

Activation of S1P1 and S1P3 receptors in vascular endothelia, often due to release

of S1P from activated platelets, decreases paracellular permeability due to assembly of

the circumferential actin cytoskeleton and adherens junctions [14, 106, 134]. Thus, in the

present study we examined filamentous actin, VE-cadherin, β-catenin and

phosphotyrosine at cell-cell borders of the inner wall of SC following S1P treatment. We

chose to study adherens junction proteins because they appear integral to the

Page 62: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

61 

 

development and maintenance of endothelial barrier properties [135-138]. While

cadherins directly mediate cell-cell interactions, β-catenin regulates the association of the

cadherin junction complex with the circumferential actin cytoskeleton. For example,

phosphorylation of catenins modulates the adhesive function of the cadherin junction

complex by decreasing the affinity of a cadherin for an adjacent cadherin or the affinity

of the cadherin/catenin complex for the circumferential actin cytoskeleton [139-142]. In

vascular endothelia, PECAM-1 (CD31) localizes at lateral cell borders and associates

with adherens junctions. We examined phosphotyrosine staining patterns due to two

recent reports showing modulation of PECAM-1 phosphorylation by S1P and mechanical

stress, which correlated with decreased endothelial monolayer permeability [143, 144].

The fact that we were unable to discern consistent changes in abundance or

localization of junctional proteins suggests that junctional complexity is already high

under basal conditions, and/or that additional increases in junctional assembly were not

resolvable using the confocal microscopy techniques used in the present study. It may be

easier to notice disassembly of junctions between SC cells due to appearance of pores or

gaps in staining. Alternatively, it may be that consistent changes were not observed

because it is estimated that in older eyes only a fraction of the total circumference of the

conventional outflow pathway typically conducts flow [145]. Since flow pathways were

not “marked” with tracer in the present study, only a subset of the eye regions that were

examined by microscopy will have been exposed to S1P; this may partially explain the

inconsistent changes we observed with filamentous actin staining.

Page 63: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

62 

 

We and Mettu et al. did not notice a change in the morphology of the TM

following S1P treatment. While the TM probably participates in the S1P effects,

morphological alterations in the TM were not discernable due to the limitation of the

methods used in our study and the previous one. If cellular contraction occurs in the TM

following exposure to S1P, then other endpoints, such as myosin light chain

phosphorylation status, or filamentous to globular actin ratio in the TM may represent

better ways to monitor change in TM contractility.

As a constituent of aqueous humor and released factor from activated platelets in

the SC lumen, S1P is expected to contribute to a tonal level of signaling and tissue

homeostasis in the conventional outflow pathway [146]. Thus, a potential therapeutic

strategy to treat those with ocular hypertension is the development of S1P receptor

antagonists, which should decrease S1P-mediated tone and hence increase outflow

facility. The relative contribution of S1P receptor subtypes expressed by outflow cells

will dictate whether specific- or pan-antagonists to the S1P1 and S1P3 receptors would be

more efficacious.

Page 64: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

63 

 

4. SPHINGOSINE-1-PHOSPHATE ENHANCEMENT OF CORTICAL

ACTOMYOSIN ORGANIZATION IN CULTURED HUMAN

SCHLEMM’S CANAL ENDOTHELIAL CELL MONOLAYERS

This chapter has been published in Investigative Ophthalmology and Visual

Science: Sumida, G.M. and Stamer, W.D. (2010). Sphingosine-1-phosphate enhancement

of cortical actomyosin organization in cultured human Schlemm's canal endothelial cell

monolayers. Invest Ophthalmol Vis Sci. 2010 June 30 [Epub ahead of print].

4.1 Introduction

Sphingosine-1-phosphate (S1P) is an endogenous bioactive lipid implicated in

many systemic processes, such as bradycardia, vasoconstriction, angiogenesis, and

lymphocyte egress [11, 12]. S1P binds with high affinity to five different G-protein

coupled receptor subtypes (S1P1-5), formerly known as EDG receptors [81]. Found in the

nanomolar range in plasma with the majority bound to high-density lipoproteins [70],

S1P has also been detected in aqueous humor [125]. Perfusion of S1P in human and

porcine whole-eye organ culture models reduces outflow facility, thereby increasing

resistance in the conventional outflow pathway [9, 147]. The majority of resistance to

aqueous humor outflow is attributed to the juxtacanalicular region of the TM and the

inner wall of SC [5-8]. Histological examination of S1P-treated and control porcine eyes

revealed no differences in the structure of the TM [9]. On the other hand, S1P-treated

Page 65: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

64 

 

eyes displayed a dramatic increase in giant vacuoles along the endothelial lining of the

aqueous plexus (analogous to SC in humans) compared to untreated control in eyes fixed

at the same pressure. Giant vacuoles form in response to a pressure-drop across the

endothelial lining, indicating a potential enhancement of endothelial cell-cell junction

complexity and an increase in resistance to outflow following activation of S1P receptors

along the endothelial lining. Activation of S1P receptors expressed by inner wall of SC

may also explain dramatic effects of S1P on resistance to outflow observed in humans

[147].

In support of S1P affecting the junctions between SC cells lining the inner wall,

vascular endothelial cells treated with S1P increase junctional proteins to sites of cell-cell

contact [14-16]. Additionally, S1P promotes cortical actin formation with an associated

increase in MLC phosphorylation at the cell cortex [17, 19]. This actomyosin

organization provides stability for the cell-cell and cell-matrix junctions and is essential

in maintaining endothelial barrier function. On the other hand, MLC phosphorylation in

S1P-treated contractile cells (like TM cells) occurs with focal adhesion and stress fiber

formations [9]. While the contractile effects observed in TM cells involve the small

GTPase Rho [9], the S1P effects observed in endothelial cells are conferred by the small

GTPase Rac [148]. Both Rac and Rho affect cytoskeleton dynamics and intercellular

junctions and can either increase (Rac) or decrease (Rho) endothelial barrier function

[13].

While S1P’s effects on human TM have been investigated, the mechanism by

which S1P works through SC cells has not. The present study was performed to test the

Page 66: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

65 

 

hypothesis that SC cells respond to S1P in culture by activating Rac and increasing

actomyosin reorganization at the cell cortex. To accomplish this objective, we monitored

the extent of RhoA versus Rac activation, and MLC phosphorylation in S1P-treated

human SC cells in culture compared to control. Additionally, we examined phospho-

MLC, F-actin, and β–catenin localization to determine S1P involvement in cytoskeletal

organization and mobilization of cell junction proteins in SC cells.

Page 67: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

66 

 

4.2 Methods

4.2.1 Cell Culture

Human SC and TM cells were isolated from donor eyes using methods previously

described [126, 127]. Both cell types were maintained in Dulbecco’s modified Eagle’s

medium (DMEM, low glucose, Invitrogen) supplemented with 10% fetal bovine serum,

penicillin (100 units/ml), streptomycin (0.1 mg/ml), and glutamine (0.29 mg/ml). At

least 2 different cell strains isolated from two different donor eyes were tested in each

type of experiment. The cell strains chosen for each experiment were based on

availability at the time of the experiments. Human umbilical vein endothelial cells

(HUVEC-2, BD Biosciences, Bedford, MA) were maintained in Medium 199

(Invitrogen) supplemented with 15% fetal bovine serum, heparin sodium salt (90 μg/ml,

Sigma-Aldrich, St. Louis, MO), endothelial mitogen (0.1 mg/ml, Biomedical

Technologies Inc., Stoughton, MA), penicillin (100 units/ml), streptomycin (0.1 mg/ml),

and glutamine (0.29 mg/ml).

4.2.2 Immunoblot analyses

Cells, following 2 hr serum starvation and appropriate treatments, were washed

twice with ice-cold PBS, lysed with ice-cold Laemmli buffer, and boiled for 5 min. The

samples were then separated on a 12% polyacrylamide gel by SDS-PAGE and

electrophoretically transferred to nitrocellulose membranes for 90 min at 100 volts. The

membranes were blocked with 5% milk in Tris-buffered saline containing 0.2% Tween-

20 (TBS-T) for 1 hr, then incubated overnight at 4ºC with rabbit IgGs against myosin

Page 68: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

67 

 

light chain or phosphor (Thr18/Ser19)-myosin light chain (1:1000 dilution, Cell

Signaling, Beverly, MA). Following exposure to primary antibody, membranes were

washed with TBS-T, incubated with horseradish peroxidase-conjugated goat anti-rabbit

or goat anti-mouse IgGs (40 ng/ml, Jackson Immunoresearch Laboratories, West Grove,

PA) for 1 hr at room temperature, then washed again with TBS-T (3 x 10 min).

Membranes were incubated with either Amersham ECL Advance (GE Healthcare) or

HyGLO (Denville Scientific, Metuchen, NJ) chemiluminescence reagents and exposed to

X-ray film (Genesee Scientific, San Diego, CA). Membranes were re-probed with ascites

fluid containing mouse monoclonal IgG against β-actin (1:10,000 dilution, Sigma-

Aldrich) for loading control. Signals in the linear range of the X-ray film were captured

digitally and densitometry was performed using a gel documentation and analysis system

(GeneSnap and GeneTools software, Syngene, Frederick, MD).

4.2.3 RhoA activation assay

Active RhoA was measured using a RhoA activation assay kit (Millipore). The

assay is based on affinity precipitations previously described to pull-down GTP-bound

Rho [149]. In brief, cells in a 6-well plate at confluence for at least one week were serum

starved for 2 hr prior to treatment. Following treatment, cells were washed twice with

ice-cold PBS, lysed with ice-cold magnesium lysis buffer (MLB), then clarified by

centrifugation at 14,000 x g at 4°C for 5 min. Cell lysates were then incubated with 7.5

μg of Rho assay reagent slurry (glutathione-agarose with GST-tagged Rhotekin Rho

binding domain) for 45 min at 4°C. Additional cell lysates were loaded with GDP or

Page 69: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

68 

 

GTPγS prior to pull-down to serve as negative and positive controls, respectively.

Following pull-downs, beads were washed three times with MLB. Laemmli sample

buffer was then added to the samples and boiled for 5 min. GTP-bound RhoA was

detected via immunoblot analysis using a mouse monoclonal antibody against RhoA

(26C4, 1 μg/ml, Santa Cruz Biotechnology, Santa Cruz, CA). A sample from each cell

lysate was used to probe for total RhoA.

4.2.4 Rac activation assay

The colorimetric Rac G-Lisa assay kit (Cytoskeleton, Denver, CO) was used to

measure Rac (1,2,3) activation. Upon reaching confluence, cells were serum starved for 2

hr prior to treatments. Following treatments, cells were washed once with ice-cold PBS,

lysed, and clarified by centrifugation at 14,000 rpm at 4°C for 2 min. An aliquot from

each sample was used to measure and normalize protein concentrations between the

samples. 50 µl of each lysate was added to separate wells of the plate coated with the

Rac-GTP binding domain of p21 activated kinase. Additional wells were filled with lysis

buffer or non-hydrolyzable Rac to serve as a negative and positive control, respectively.

The plate was placed immediately on an orbital shaker set at 200 rpm for 30 min at 4°C.

The plate was then washed three times with wash buffer at room temperature, incubated

with 200 µl of antigen presenting buffer for 2 min, and washed three more times. Mouse

monoclonal IgG against Rac (1:200 dilution) was added to each well and incubated for 45

min on the orbital microplate shaker at room temperature. After three more washes,

secondary anti-horseradish peroxidase (HRP)-labeled antibody (1:100 dilution) was

Page 70: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

69 

 

added to each well and incubated on the orbital microplate shaker for an additional 45

min. The plate was washed three times, then 50 µl of HRP detection reagent was added

and incubated at 37°C for 5 min. HRP stop buffer (50 µl) was added, and the absorbance

readings at 490 nm were measured using the Flexstation 3 plate reader (Molecular

Devices, Sunnyvale, CA). The mean Abs490 values acquired for each treatment were

compared to the control and analyzed by a two-tailed, unpaired student’s t-test.

Differences were considered significant at p<0.05.

4.2.5 Immunofluorescence microscopy

Cells plated on glass coverslips were maintained at confluence for at least a week

and then serum starved for 2 hr prior to treatments. Following treatments, cells were

washed with ice-cold PBS, fixed with 4% paraformaldehyde for 10 min, permeabilized

with PBS containing 0.2% Triton X-100 (PBS-T) for 5 min, and blocked with 10% goat

serum in PBS-T for 30 min. After blocking, the cells were incubated for 1 hr at room

temperature with rabbit IgGs against either phospho-specific (Ser19) myosin light chain

(2.5 μg/ml, Millipore), β-catenin (1 μg/ml, Santa Cruz Biotechnology), or cortactin

(1:100 dilution, Cell Signaling). The cells then underwent three washes with PBS, 1 hr

incubation with CY3-conjugated goat anti-rabbit IgG (0.75 μg/ml, Jackson

Immunoresearch Laboratories), one wash with PBS, counterstain with SYTOX green

nucleic acid stain (100 nM, Invitrogen) for 1 min, and three more washes with PBS

before visualization. Background and auto-fluorescence was detected by incubating

CY3-conjugated goat anti-rabbit IgG without previous primary antibody incubations. To

Page 71: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

70 

 

detect F-actin, confluent cultures of cells were stained with AlexaFluor 568 phalloidin

(Invitrogen) for 20 min. The labeled cells were visualized and captured digitally using a

Nikon PCM 2000 confocal microscope (Melville, NY).

Page 72: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

71 

 

4.3 Results

To study the effects of S1P on cultured SC cell monolayers, we first investigated the

Rho/Rho-kinase/MLC phosphorylation pathway that is activated by S1P in TM cells [9].

Stimulation of this pathway alters the contractile state of cells by modifying actomyosin

architecture. Using a pull-down assay to detect GTP-bound (active) RhoA, 1 μM S1P

treatments increased activated RhoA in a time-dependent manner compared to untreated

control (Figure 4.1).

Figure 4.1. S1P-induced RhoA activation and MLC phosphorylation in SC cell monolayers. (A) RhoA activation (RhoA-GTP) in SC cells treated with 1 μM S1P for 5 and 30 min were compared to control (C). Cell lysates were also probed for total RhoA and used for comparison between the treatment groups. Blots are representative of 4 different experiments using 2 SC cell strains. (B) TM and SC cells were treated with 1 μM S1P, or left untreated (C), and probed for phospho-MLC over a time course (1, 3, 5, 30, and 60 min) reflecting the S1P effect in organ-culture. β-actin was used as a loading control. The TM blot is representative of 7 experiments using 3 cells strains while the SC blot is representative of 8 experiments using 3 cell strains.

Page 73: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

72 

 

Figure 4.2. Dose-dependent S1P-mediated MLC phosphorylation in both TM and SC cell monolayers. SC and TM cells were treated with a range (10-4 to10-8 M) of S1P concentrations for 5 min. Graphs represent the percent MLC phosphorylation observed (phospho-MLC/β-actin) in comparison to the maximum response of each experiment (mean ± SEM). (A) SC cells displayed an EC50 value of 0.83 μM (n=7), while (B) TM cells displayed a value of 1.33 μM (n=5).

Downstream of activated Rho, Rho-kinase phosphorylates and deactivates MLC

phosphatase, thus allowing an increase in phospho-MLC species [150]. Corresponding to

the acute effect of S1P in organ culture [9, 147], we examined effects of 1 μM S1P on

MLC phosphorylation in TM and SC cell monolayers between 1-60 minutes. For both

TM and SC cells, maximal MLC phosphorylation levels occurred in the 5 to 30 min time

frame (Figure 4.1). No significant differences in β-actin levels were observed between

the different time treatments. With similar time-dependent increases in phospho-MLC,

we next investigated S1P dose-related MLC phosphorylation in both SC and TM cells

Page 74: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

73 

 

over a range of S1P concentrations (10-4 to10-8 M) at the 5 min time point. We observed

that SC and TM cells (Figure 4.2) dose-dependently increased phospho-MLC with

increasing S1P concentrations. Maximal phospho-MLC levels in both cell types were

approached with 10-100 μM S1P with EC50 values of 0.83 μM and 1.33 μM for SC and

TM cells, respectively.

Figure 4.3. Rho-kinase dependent MLC phosphorylation by S1P in SC cell monolayers. TM and SC cells were treated with 1 unit/ml thrombin (T), 1µM S1P (S), or left untreated (C) for 5 min. 10 µM Y-27632 (Y) was pre-incubated for 30 min prior to thrombin or S1P treatments. Two exposure timepoints (exposure 1 and exposure 2) of blots on film are displayed to compare phospho-MLC levels between TM and SC with β-actin used as a loading control. Blots are representative of at least 6 experiments.

A 30 min pre-incubation with the Rho-kinase inhibitor Y-27632 blocked S1P-

induced MLC phosphorylation in SC cells similarly to what is observed in TM cells,

indicative of S1P activation of the RhoA/Rho-kinase/MLC phosphorylation pathway

(Figure 4.3). Y-27632 also inhibited MLC phosphorylation by thrombin, a known

activator of the same pathway.

Upon examining these two cell types more closely, we found that TM cells

contained a greater abundance of phospho-MLC species compared to SC cells following

Page 75: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

74 

 

S1P treatment (Figure 4.3). Comparisons were made in experiments where attention was

paid to equal protein loading of SC and TM cell lysates and differences were revealed

after analysis of two different time exposures of X-ray film exposed to a single blot

(Figure 4.3). Likewise, a greater expression level of MLC was observed in the TM cell

strains compared to SC (Figure 4.4) in all three cell strains of each cell type tested.

Figure 4.4. MLC expression levels in TM and SC cell monolayers. Whole-cell lysates from 3 TM (86, 90, 93) and 3 SC (51, 53, 56) cell strains were probed for MLC. Two exposure time points (exposure 1 and exposure 2) of blots on film are displayed to distinguish MLC levels between and within each cell type with β-actin used as a loading control.

S1P-mediated Rac activation in endothelial cells promotes barrier integrity and

decreased paracellular permeability [13]. Since S1P receptors in endothelia often

preferentially utilize the Rac pathway, we next investigated whether S1P receptors in SC

endothelial cells signal through the Rac pathway. In fact, compared to untreated control

(0.20 ± 0.04), 1 μM S1P treatments for 5 min (0.36 ± 0.07) and 30 min (0.29 ± 0.03)

significantly increased Rac (1,2,3) activation (mean Abs490 ± SD, p<0.01) (Figure 4.5) in

mature SC monolayers.

Page 76: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

75 

 

Figure 4.5. S1P-induced Rac activation in SC cell monolayers. Rac (1,2,3) activation in SC cells treated with 1 μM S1P for 5 and 30 min were compared to control. The graph represents the mean Abs490 ± SD observed in 3 different cell strains (n=6, *p<0.01).

With S1P-induced Rho and Rac activation known to affect cytoskeletal

organization and endothelial permeability [13, 103], we next examined the effect of S1P

on F-actin arrangement and phospho-MLC localization. In SC cells, S1P decreased stress

fiber polymerization and increased cortical actin assembly compared to control following

5 and 30 min treatments (Figure 4.6). Likewise, phospho-MLC staining was detected in

a cortical arrangement following S1P treatment (Figure 4.6). The cortical staining pattern

is similar to what was observed in HUVEC cells (Figure 4.7). An increase in global

phospho-MLC staining in S1P-treated over control cells was consistent with the increase

in phospho-MLC observed through immunoblot analysis (Figure 4.2).

Page 77: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

76 

 

Figure 4.6. S1P promotes cortical actin assembly and peripheral localization of phospho-MLC localization in SC monolayers. SC cells were treated with 1 μM S1P for 5 and 30 min, or left untreated (control). (A) Cells were stained with AlexaFluor phalloidin 568; arrows indicate cortical actin while arrowheads indicate stress fiber formations. Results shown are representative of 7 experiments with 2 different cell strains. (B) In parallel experiments, cells were also immunolabeled for phospho-MLC (red) and counterstained with SYTOX green to identify nuclei (green). Arrows display cortical arrangement of phospho-MLC. The inset in the control panel displays secondary antibody staining alone (2ºAb), while the inset in the S1P 30’ panel is an enlargement focusing on cortical phospho-MLC staining. Results shown are representative of 4 experiments with 3 different cell strains.

Page 78: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

77 

 

Figure 4.7. S1P and phospho-MLC localization in HUVEC monolayers. Human umbilical vein endothelial cells (HUVEC) were immunolabeled for phospho-MLC (red) and counterstained with SYTOX green to identify nuclei (green). Arrows display cortical arrangement of phospho-MLC. The inset in the control panel displays secondary antibody staining alone (2ºAb).

Figure 4.8. S1P and β-catenin localization in SC. Monolayers of SC cells were treated with 1 μM S1P for 30 min or left untreated. Cells were immunolabeled for β-catenin (red) and counterstained with SYTOX green to identify nuclei (green). Arrows display β-catenin at the cell periphery. The inset displays secondary antibody staining alone (2ºAb). Results shown are representative of 5 experiments with 3 different cell strains.

Page 79: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

78 

 

Following the observation of increased cortical actin and phospho-MLC

following S1P treatment, we next investigated whether β-catenin is also recruited to the

cell periphery. β-catenin is an adaptor protein associated with adherens junctions and has

been shown to increase localization to sites of cell-cell contact in vascular endothelial

cells [15]. Indeed, S1P treatment increased β-catenin recruitment to the cell periphery of

SC cell monolayers (Figure 4.8).

Page 80: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

79 

 

4.4 Discussion

The major findings of this study are that human SC cell monolayers in culture

respond to S1P by activating both RhoA and Rac. Stimulation of these small GTPases

results in a coordinated increase in MLC phosphorylation, F-actin, and β-catenin at the

cell cortex. These results suggest that the endothelial cells lining the inner wall of SC

contribute to the S1P effects on outflow facility through actomyosin organization at the

cell cortex and enhancing cell-cell junction assembly, thereby increasing trans-

endothelial resistance.

We observed that S1P-treated SC cells respond in part via RhoA activation and a

Rho-kinase-dependent MLC phosphorylation. In addition, SC cells increase MLC

phosphorylation in a similar time and dose-dependent manner with TM cells. Although

S1P activates the RhoA-MLC signaling pathway in both cell types, the greater MLC

expression levels and larger extent of MLC phosphorylation in S1P-treated TM cells

indicates that the Rho pathway is likely more dominant in TM compared to SC. These

data are in agreement with S1P increasing stress fiber formation in TM cells [9] and TM

behavior as a contractile tissue.

Despite the activation of RhoA and subsequent MLC phosphorylation in S1P-

treated SC cells, the peripheral localization of phospho-MLC and concurrent cortical

actin formation suggests the strengthening of the cytoskeletal network at the cell cortex.

Hence, actomyosin reorganization in S1P-stimulated endothelial cells is linked with an

increase in trans-endothelial electrical resistance and cell barrier integrity [17], two

parameters that are dependent upon Rac activation and adherens junction assembly [13,

Page 81: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

80 

 

15]. When SC cells were tested in the present study, S1P robustly increased Rac activity

and mobilization of the adherens adaptor protein, β-catenin. Taken together, SC cells

appear unique in that S1P signals effectively through both Rac and RhoA compared to

their endothelia brethren; likely due to SC’s unusual requirement to withstand fluid flow

in the basal to apical direction (compared to apical to basal for nearly all other

endothelia). Thus for tissue homeostasis, the inner wall of SC must not only rely on cell-

cell attachments to maintain a patent monolayer, but also on unusually strong cell-matrix

attachments through focal adhesions, secured intracellularly by actin stress fibers.

While the S1P effects on the actomyosin architecture in SC and TM may differ, a

combination of those responses may be essential towards decreasing outflow facility in

situ. In TM cells, a Rho-dominant response to S1P leads to elevated MLC

phosphorylation with the assembly of stress fibers. In SC cells, a moderate increase in

Rho activity and MLC phosphorylation leads to stress fiber assembly. In parallel an

increase in Rac activity, leads to cortical actomyosin arrangement (See Figure 4.9).

Thus, simultaneous TM cell contraction and an increase in SC endothelial barrier

integrity with a decrease in aqueous humor flow across the inner wall may underlie S1P’s

dramatic and immediate effect on outflow facility.

Our observed change in the distribution of β-catenin following S1P treatment of

cultured SC cells in this study is similar to findings in some, but not all areas of the inner

wall examined in a recent in situ [147] study by our group. One possible explanation for

this inconsistency is segmental flow of aqueous humor through the conventional outflow

pathway [145]. With segmental flow, only a fraction of the conventional outflow tissues

Page 82: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

81 

 

(including the inner wall) encounter flow (and thus S1P), making interpretation of data

obtained from areas of inner wall where exposure to segmental flow is unknown very

difficult.

Figure 4.9. Model for differential S1P receptor coupling to Rac/Rho pathways in outflow cells. The Rho pathway is preferentially activated by S1P in TM cells and promotes stress fiber formation and cell contractility while the Rac pathway is preferentially activated in vascular endothelial cells (VEC) and promotes cortical actin formation with an increase in endothelial barrier function. In SC cells, both Rho and Rac are activated by S1P with a transition from stress fibers to cortical actin formation.

With the establishment of S1P’s effects in both organ-culture and cell-based

assays, we can now utilize subtype-specific antagonists to identify the receptors

responsible for each response. Binding of an antagonist to the S1P receptor subtype

responsible for increasing outflow resistance may inhibit endogenous tone, decrease

outflow resistance and thus provide a novel drug target for primary open-angle glaucoma

Page 83: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

82 

 

to reduce ocular hypertension. Interestingly, intravenous injection of an S1P1-specific

antagonist in mice increases lung capillary leakage [121]. Here, the S1P1 receptor is

linked to maintaining endothelial barrier integrity through mechanisms involving Rac

activation, cytoskeletal rearrangement, and junctional organization [148]. Thus, we

predict that a S1P1 antagonist-induced decrease in basal S1P1 receptor activity in SC cells

may decrease outflow resistance, thereby increasing aqueous humor outflow.

Page 84: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

83 

 

5. S1P2 RECEPTOR REGULATION OF SPHINGOSINE-1-PHOSPHATE

EFFECTS ON CONVENTIONAL OUTFLOW PHYSIOLOGY

5.1 Introduction

Elevated intraocular pressure (IOP), a major risk-factor for glaucoma, is due to an

increased resistance to aqueous humor outflow in the pressure-responsive, conventional

pathway. The majority of resistance to outflow is attributed to the juxtacanalicular

connective tissue (JCT) region of the trabecular meshwork (TM) and the inner wall of

Schlemm’s canal (SC) [5-8]. To enter the SC lumen and return to venous circulation,

aqueous humor flows through the JCT, a 2-20 µm thick region adjacent to SC that is

comprised of a loose arrangement of extracellular matrix interspersed with TM cells that

are attached to SC cells and the basal lamina [43]. Aqueous humor then crosses the inner

wall of SC, a continuous endothelial monolayer. It is important to determine how these

tissues regulate resistance in order to find new mechanisms to lower IOP in those with

glaucoma.

The endogenous bioactive lipid sphingosine-1-phosphate (S1P) decreases outflow

facility, thereby increasing resistance, in both porcine and human eyes [9, 147].

Originally believed to be an intracellular signaling molecule, S1P is now known to be

involved in receptor-mediated signaling following the identification of S1P as the ligand

for the EDG1 receptor (S1P1) [10]. Of the five S1P receptors (S1P1-5) that have been

identified, the S1P1-3 receptors are ubiquitously expressed, while S1P4 is limited to

Page 85: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

84 

 

lymphoid and S1P5 to brain and skin tissue [12]. The regulation of cytoskeletal

dynamics is a main downstream effect of S1P receptor activation [13]. For example, in

cultured TM cells, S1P induces Rho activation, thus promoting myosin light chain (MLC)

phosphorylation and increased stress fiber formation [9]. In SC cells, S1P increases

MLC phosphorylation while also promoting a cortical rearrangement of the actomyosin

system [151]. The cortical actomyosin assembly in SC cells is similar to the S1P effect

observed in vascular endothelial cells, an effect mediated by the S1P1 receptor to promote

endothelial barrier function [148].

The S1P receptor subtype(s) that is responsible for mediating the decrease in

outflow facility by S1P represents a viable pharmacological target to modulate outflow

resistance and reduce IOP. Therefore, the aim of this study was to identify S1P

receptor(s) that mediate effects on outflow facility by testing pharmacology of receptor-

selective compounds in the porcine and human whole eye perfusion models, as well in

human primary cultures of SC and TM cells. Despite similarities between SC and

vascular blood endothelia [20] and the robust expression of the S1P1 receptor in SC cells

[147], the S1P1-specific agonist SEW2871 [152] failed to decrease outflow facility in the

human whole-eye perfusion and promote MLC phosphorylation in both SC and TM cells.

Additionally, the antagonists W146 (S1P1-specific) [121] and VPC23019 (S1P1,3-

specific) [153] failed to block the S1P increase in MLC phosphorylation. However, the

S1P2-specific antagonist JTE-013 [122] blocked the S1P decrease of outflow facility in

both porcine and human whole-eye perfusions, as well as the S1P-promoted MLC

phosphorylation in SC and TM cells.

Page 86: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

85 

 

5.2 Methods

5.2.1 Reagents

All chemicals and reagents used were purchased from Sigma-Aldrich (St. Louis,

MO) unless otherwise noted. S1P was dissolved by adding to methanol and boiled.

Methanol was then evaporated with a stream of nitrogen gas. The subsequent thin film of

S1P was solubilized in phosphate-buffered saline (PBS) containing 4 mg/ml fatty acid-

free bovine serum albumin. W146 and VPC23019 were acquired from Avanti Polar

Lipids (Alabaster, AL) while SEW2871 and JTE-013 were acquired from Cayman

Chemical (Ann Arbor, MI).

5.2.2 Eye tissue

Enucleated human donor eyes were obtained from the National Disease Research

Interchange (Philadelphia, PA), Sun Health Research Institute (Sun City, AZ), and Life

Legacy Foundation (Tucson, AZ). The enucleated human eyes were free of any known

ocular disease and stored in a moist chamber at 4ºC until use. Enucleated porcine eyes

were obtained from the University of Arizona Meat Sciences Laboratory.

5.2.3 Cell culture

Human SC cells were isolated from donor eyes using a cannulation technique, and

characterized and cultured as previously described [126]. Human TM cells were isolated

using a blunt dissection technique, followed by extracellular matrix digestion.

Characterization and culturing were performed as previously described [127]. Both cell

Page 87: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

86 

 

types were maintained in Dulbecco’s modified Eagle’s medium (DMEM, low glucose)

supplemented with 10% fetal bovine serum, penicillin (100 units/ml), streptomycin (0.1

mg/ml), and glutamine (0.29 mg/ml). The SC cell strains used were SC51, SC53,

SC55/56, and SC58 that were derived from donors of ages 66, 44, 29, and 34 years,

respectively. The TM cell strains used were TM86, TM88, TM90, and TM 93 that were

derived from donors of ages 3 months, 25 years, 4 months, and 35 years, respectively.

At least 2 different cell strains were tested in each type of experiment and were chosen

based on strain availability at the time of the experiments.

Human umbilical vein endothelial cells (HUVEC-2, BD Biosciences, Bedford,

MA) were maintained in Medium 199 (Invitrogen) supplemented with 15% fetal bovine

serum, heparin sodium salt (90 μg/ml), endothelial mitogen (0.1 mg/ml, Biomedical

Technologies Inc., Stoughton, MA), penicillin (100 units/ml), streptomycin (0.1 mg/ml),

and glutamine (0.29 mg/ml).

5.2.4 Immunoblot analyses

Immunoblot analyses were performed as previously described [151].

Nitrocellulose membranes were blocked with 5% milk for 1 hr, then incubated overnight

at 4ºC with rabbit IgGs against phospho (Thr18/Ser19)-myosin light chain (1:1000

dilution, Cell Signaling, Beverly, MA), phospho (Ser473)-Akt (1:2000 dilution, D9E,

Cell Signaling), or pan-Akt (1:1000 dilution, C67E7, Cell Signaling). Secondary

antibodies used were horseradish peroxidase-conjugated goat anti-rabbit or goat anti-

mouse IgGs (40 ng/ml, Jackson Immunoresearch Laboratories, West Grove, PA) for 1 hr

Page 88: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

87 

 

at room temperature. Membranes were incubated with either Amersham ECL Advance

(GE Healthcare) or HyGLO (Denville Scientific, Metuchen, NJ) chemiluminescence

reagents and exposed to X-ray film (Genesee Scientific, San Diego, CA). Membranes

were re-probed with ascites fluid containing mouse monoclonal IgG against β-actin

(1:10,000 dilution, Sigma-Aldrich, St. Louis, MO) for a loading control. Protein signals

were captured digitally and densitometry was performed (GeneSnap and GeneTools

software, Syngene, Frederick, MD).

5.2.5 Human whole-eye perfusion

Whole-eye perfusions were performed based on the system described by Ethier et

al. [128]. Upon enucleation, eyes were stored in a moist chamber at 4ºC until the

beginning of the experiment. At the beginning of an experiment, eyes were placed in

PBS at 34ºC and a needle was inserted through the cornea and placed in the posterior

chamber, behind the iris. The eyes were then perfused with Dulbecco’s PBS containing

5.5 mM glucose (DBG) pre-filtered through a syringe-driven filter unit with 0.22 μm

pores (Millipore). Human eyes were perfused at a constant pressure of 8 mmHg

(equivalent to 15 mmHg in vivo) for 60-90 min to establish a baseline outflow facility

(stable flow for at least 30 min). Following baseline perfusion, the contents of the

anterior chamber were exchanged with DBG containing either experimental compounds,

or control. Following exchanges, perfusions at 8 mmHg were resumed and new baselines

were reached (defined as reasonably stable flow for at least 30 min prior to the end of the

perfusion). Normalized facilities were then calculated, dividing the new baseline

Page 89: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

88 

 

following exchange by the original baseline facility. Additionally, the percent net facility

change between paired eyes were calculated by subtracting the percent facility change

from baseline in the contralateral control eye from the percent facility change in the

experimental eye.

5.2.6 Porcine whole-eye perfusion

Porcine eyes were enucleated immediately following time of death (TOD). The

eyes were received within 2 hrs of TOD and used in experiments within 12 hrs of TOD.

The porcine eyes were perfused using the same procedure as the human eye perfusions,

but were perfused at a constant pressure of 15 mmHg as has been done previously [9].

The porcine eye perfusions were used to test the effects of antagonist +/- S1P. After a

stable baseline, eyes were exchanged with antagonist and perfused for an additional 1 hr

in order to expose the outflow tissue to the antagonist and determine whether facility

changes occur with antagonist alone. The eyes were then exchanged a second time with

antagonist alone or antagonist + S1P, and then perfused for an additional 2 hrs. Separate

experiments were also done to test S1P’s effect in porcine eyes without antagonists.

Control eyes received media alone in both exchanges, while experimental eyes received

media first, then S1P in the second exchange.

5.2.7 Statistical analyses

Results for human whole-eye perfusions were analyzed using a two-tailed, paired

student’s t-test. Porcine whole-eye perfusions were analyzed using a two-tailed,

Page 90: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

89 

 

unpaired student’s t-test with equal variance. Cell culture experiments were analyzed

using a one-way ANOVA with Dunnett’s post-hoc test. Results were considered

significant when p< 0.05.

Page 91: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

90 

 

5.3 Results

We used two complementary approaches, outflow facility in whole-eye perfusions and

MLC phosphorylation in cultured cells, to determine whether the S1P1 receptor is

responsible for S1P effects on outflow resistance in human eyes. In human whole-eye

perfusions with SEW2871, the mean donor age of perfused eyes was 82.7 years (range

Table 5.1. Donor information and summary of results from human whole-eye perfusions

Outflow Facility (μl/min/mmHg) % facility % net facility

ID Age Gender TOD-TOE TOD-TOP Stable baseline New baseline change change

SEW2871 63E 77 F 4.5 9.5 0.220 0.246 11.8 10.1

64C 0.119 0.121 1.7

67C 84 F 3 29.5 0.192 0.170 -11.5 -5.2

68E 0.240 0.200 -16.7 69C 87 M 2 13.5 0.101 0.118 16.8 -17.8

70E 0.196 0.194 -1.0

Mean 82.7 2F/1M 3.2 17.5 Control 0.137 ± 0.048 0.136 ± 0.029 2.4 ± 14.2 -4.3 ± 14.0

Experimental (5 μM SEW2871) 0.219 ± 0.022 0.213 ± 0.028 -2.0 ± 14.3

JTE-013 + S1P 87C 85 M 2.6 12.5 0.164 0.121 -26.2 23.8

88E 0.168 0.164 -2.4

89E 82 M 2.4 23 0.200 0.218 9.0 34.6

90C 0.227 0.169 -25.6

91C 80 F 9 22.5 0.239 0.192 -19.7 -2.9

92E 0.235 0.182 -22.6

101E 62 M 4 15 0.106 0.173 63.2 80.6

102C 0.242 0.200 -17.4

103C 89 M unkn 15 0.272 0.225 -17.3 22.4

104E 0.255 0.268 5.1 117C 84 F 2 10 0.186 0.144 -22.6 27.3

118E 0.190 0.199 4.7 Mean 80.3 2F/4M 4.0 16.3 Control (5 μM S1P) 0.222 ± 0.040 0.175 ± 0.038 -21.4 ± 4.0 31.0 ± 27.4 Experimental (5 μM JTE-013 + 5 μM S1P) 0.192 ± 0.053 0.201 ± 0.038 9.5 ± 28.6 Control and experimental values are means ± SD; C, control; E, experimental; TOD, time of death; TOE, time of enucleation; TOP, time of perfusion; unkn, unknown

Page 92: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

91 

 

77-87) and the mean time of death to start of perfusion was 17.5 hrs (range 9.5-29.5)

(Table 5.1). Unlike the 36% ± 20% decrease in outflow facility previously observed in

S1P-treated eyes [147], 5 µM SEW2871-treated eyes displayed no differences in outflow

facility compared to contralateral control eyes up to 1hr following anterior chamber

exchanges (-4.3 ± 14.0 % net facility change, n=3). The normalized facility baselines

following exchanges in control and SEW2871-treated eyes were 1.02 ± 0.14 and 0.98 ±

0.14, respectively (mean ± SD, p=0.62).

Figure 5.1. S1P1 receptor activation does not promote MLC phosphorylation. (A) SC and (B) TM cells were treated for 5 min with S1P and the S1P1-specific agonist SEW2871 (SEW) at concentrations of 10, 1 and 0.1 μM, and then probed for MLC phosphorylation (P-MLC). β-actin was used as a loading control. Blots are representative of five experiments using two cell strains each for both cell types. (C) SEW2871 activity was confirmed through Akt phosphorylation (P-Akt) in human umbilical vein endothelial cells (HUVEC) following 5 min SEW2871 treatments at concentrations of 10, 1, and 0.1 µM. Blots were also probed for pan-Akt as a loading control. Blots are representative of four experiments.

Page 93: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

92 

 

We next treated SC and TM cells with SEW2871 to determine whether S1P1

participates in the S1P-promoted increase in MLC phosphorylation, a prominent and

reliable effect in both cell types [9, 151]. Following a 2hr serum starvation, SEW2871

(10, 1, and 0.1 µM) did not induce MLC phosphorylation in both SC and TM cells

(Figure 5.1), thus indicating that S1P1 activation alone does not promote MLC

phosphorylation. As a control, SEW2871 activity was verified with Akt phosphorylation

in human umbilical vein endothelial cells (Figure 5.1), an effect mediated by S1P1 [152].

Since S1P1 alone did not appear responsible for S1P effects on outflow resistance,

we screened three different S1P receptor-selective antagonists to test their effectiveness

in blocking the S1P –promoted MLC phosphorylation in both SC and TM cells.

Following 2 hr serum starvation, SC and TM cells were pretreated (30 min) with S1P

receptor antagonists prior to S1P treatment (5 min). In the presence of 10 μM JTE-013

(S1P2 antagonist), MLC phosphorylation levels following S1P treatments of 10, 1 and 0.1

µM were reduced compared with S1P alone in both SC and TM cells (Figure 5.2). In

agreement with the inability of SEW2871 to promote MLC phosphorylation (Figure 5.1),

the S1P1 antagonist W146, as well as the S1P1,3 antagonist VPC23019, failed to block

MLC phosphorylation by S1P.

Page 94: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

93 

 

Figure 5.2. Effect of a single concentration of S1P receptor specific antagonist on MLC phosphorylation in the presence of increasing concentrations of S1P. Cells were treated with 1 U/ml thrombin (T) and S1P at concentrations of 10, 1 and 0.1 μM for 5 min and probed for MLC phosphorylation (P-MLC). S1P effects in the presence of antagonists were tested in additional cells pre-treated with 10 μM of JTE-013 (S1P2), W146 (S1P1), or VPC23019 (S1P1,3) for 30 min. (A) SC and (B) TM blots are representative of five different experiments using three cell strains for each cell type. β-actin was used as a loading control.

Page 95: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

94 

 

Figure 5.3. Effect of increasing antagonist concentration on S1P-mediated MLC phosphorylation in SC cells. Cells were treated with 1 U/ml thrombin (T) and 1 μM S1P for 5 min and probed for MLC phosphorylation (P-MLC). S1P effects in the presence of the antagonists JTE-013 (A, S1P2 ant.), W146 (B, S1P1 ant.), or VPC23019 (C, S1P1,3 ant.) at concentrations of 10, 1 and 0.1 μM were compared. Blots are representative of six different experiments using two SC cell strains. Changes to the S1P –promoted MLC phosphorylation response (compared through the fold thrombin response) with the inclusion of antagonists are represented in the histograms beneath the correlating blots. β-actin was used as a loading control. Data are presented as mean ± SEM and are compared to S1P control response (** p<0.01, *** p<0.001).

Due to possible non-specific interactions of antagonist, we also performed the

reciprocal experiments where antagonist concentrations were varied in the presence of a

single concentration of S1P (1 µM). The 1 µM S1P-promoted MLC phosphorylation in

SC cells were significantly reduced when pretreated with all three doses of JTE-013 (10-

0.1 µM, p<0.001) (Figure 5.3). Interestingly, inclusion of 1 µM W146 reduced MLC

phosphorylation by S1P (p<0.01), although similar responses were not observed with a

higher 10 µM dose (Fig. 3B). No changes in MLC phosphorylation were observed with

the inclusion of VPC23019 in SC cells (Figure 5.3).

Page 96: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

95 

 

Figure 5.4. Effect of increasing antagonist concentration on S1P-mediated MLC phosphorylation in TM cells. Cells were treated with 1 U/ml thrombin (T) and 1 μM S1P for 5 min and probed for MLC phosphorylation (P-MLC). S1P effects in the presence of the antagonists JTE-013 (A, S1P2 ant.), W146 (B, S1P1 ant.), or VPC23019 (C, S1P1,3 ant.) at concentrations of 10, 1 and 0.1 μM were compared. Blots are representative of six different experiments using two TM cell strains. Changes to the S1P –promoted MLC phosphorylation response (compared through the fold thrombin response) with the inclusion of antagonists are represented in the histograms beneath the correlating blots. β-actin was used as a loading control. Data are presented as mean ± SEM and are compared to S1P control response (* p<0.05, ** p<0.01).

When tested in TM cells, S1P-promoted MLC phosphorylation was also

significantly reduced by JTE-013 (10 and 1 µM JTE, p<0.01; 0.1 µM JTE, p<0.05)

(Figure 5.4). The MLC phosphorylation responses by S1P were reduced with JTE-013

by 73% (10 µM), 67% (1 µM), and 57% (0.1 µM). By comparison, SC cells displayed a

more robust blocking of MLC phosphorylation at all three doses of JTE-013 by reducing

the S1P response by 83% (10 µM), 77% (1 µM), and 78% (0.1 µM). No differences in

phosphorylation status were observed with the inclusion of W146 and VPC23019 in TM

cells (Figure 5.4).

Due to the inability of the S1P1 agonist SEW2871 to mimic the S1P effects in the

whole-eye perfusion (decrease facility) and cell culture models (MLC phosphorylation),

Page 97: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

96 

 

the focus was shifted to S1P2 and the antagonist JTE-013 due to its blocking of S1P-

promoted MLC phosphorylation in our cell models. Thus, JTE-013 was included in the

whole-eye perfusion model to determine if blocking the S1P2 receptor would prevent the

S1P-induced decrease in outflow facility. In initial studies, we used porcine eyes first to

test the antagonist prior to use in human eyes due to the similar decrease in outflow

facility observed between both species [9, 147], in addition to the availability and

freshness of porcine eyes compared to human eyes.

We started with a double exchange perfusion experiment. The aim of the first

exchange was to determine if antagonist alone affects outflow while the aim of the

second exchange was to compare the inclusion of S1P with antagonist versus antagonist

alone. Parallel, control experiments using this experimental setup were also performed

(media alone for both exchanges) and experimental eyes (media in first exchange, 5 µM

S1P in second exchange), reproducing the S1P-mediated decrease in facility without

antagonist (Figure 5.5). Following verification of the S1P response in the control

experiments, experiments with the inclusion of antagonist were conducted to compare

JTE-013 control eyes (5 µM JTE-013 for both exchanges) with experimental eyes (5 µM

JTE-013 for first exchange, 5 µM JTE-013 + 5 µM S1P for second exchange). The

inclusion of antagonist, JTE-013, clearly blocks the S1P-mediated decrease in outflow

facility (Figure 5.5).

Page 98: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

97 

 

Figure 5.5. Blocking S1P2 prevents the S1P-mediated decrease in outflow facility in porcine eyes. Following a baseline perfusion, porcine eyes received two exchanges with subsequent perfusions over the course of an experiment. The 1st exchange was used to pre-treat the outflow tissue with the S1P2 antagonist JTE-013 while the 2nd exchange was used to compare eyes treated with JTE-013 alone , or in addition to 5 μM S1P (n=3). Separate control experiments using media in the 1st exchange, and in the 2nd exchange, comparing media against 5 μM S1P (n=3) were also done. Examples of outflow facility traces for experiments comparing media alone (―) to S1P (…..) (A), and comparing JTE-013 (―) to JTE-013 + S1P (…..) (B) are shown. Average normalized facilities following anterior chamber exchanges are also shown (C, D). Control and experimental perfusion data were compared following each exchange and are presented as mean ± SD (* p<0.05).

Page 99: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

98 

 

In analyzing the control experiments without antagonist (Figure 5.5), normalized

facilities following the first exchange and 1 hr perfusion for the media control and

experimental groups were 1.09 ± 0.16 and 1.12 ± 0.11, respectively (mean ± SD, p=0.76).

Following the second exchange and subsequent 2 hr perfusion, experimental eyes with

S1P displayed a typical decrease in normalized facility of 0.69 ± 0.18 compared to 1.10 ±

0.13 in the control group with media alone (mean ± SD, p=0.03, n=3) (Figure 5.5).

In the experiments with the inclusion of S1P2 antagonist (Figure 5.5), normalized

facilities following the first exchange and perfusion for the JTE-013 control and

experimental groups were 1.28 ± 0.30 and 1.21 ± 0.14, respectively (mean ± SD, p=0.74,

n=3). Following the second exchange and subsequent perfusion, normalized facilities for

the experimental eyes with JTE-013 + S1P and JTE-013 control eyes remained

comparable at 1.22 ± 0.21 and 1.36 ± 0.30, respectively (mean ± SD, p=0.53) (Figure

5.5). Thus, inclusion of JTE-013 in porcine whole-eye perfusions prevented the S1P-

induced decrease in outflow facility.

While investigating the effect of antagonist alone, normalized facilities following

the first exchange for all eyes receiving 5 μM JTE-013 (1.25 ± 0.21) displayed a trend

towards increasing outflow facility compared to all eyes receiving media alone (1.11 ±

0.13) (mean ± SD, p=0.19, n=6).

Since JTE-013 blocked the S1P-induced decrease in outflow facility in porcine

eyes, the antagonist was next used in human eye perfusions. The mean donor age for

perfused eyes was 80.3 years (range 62-89) and the mean time of death to start of

perfusion was 16.3 hrs (range 10-23) (Table 5.1). Following an initial baseline

Page 100: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

99 

 

perfusion, control eyes were exchanged with media containing 5 μM S1P while the

contralateral experimental eyes were exchanged with media containing 5 μM JTE-013 +

5 μM S1P. Due to limitations in the freshness of human tissue post-mortem, JTE-013

and S1P were given together in one exchange. After 2 hrs of perfusion following the

exchange, JTE-013 + S1P eyes and S1P control eyes displayed normalized facilities of

1.10 ± 0.29 and 0.78 ± 0.04, respectively (mean ± SD, p<0.05, n=6) (Figure 5.6). The net

facility difference with the addition of JTE-013 was 31.0 ± 27.4 %. Interestingly, the

inclusion of JTE-013 in human eyes increased outflow facility above baseline despite the

presence of S1P. Even though the antagonist appeared not to have an effect in one eye

pair (91C, 92E), JTE-013 still significantly blocked the S1P decrease of outflow facility

in human eyes.

Figure 5.6. Outflow facility effects of S1P2 receptor blockade in S1P-perfused human eyes. Following a baseline perfusion in paired human eyes, the contents of the anterior chambers of the experimental eye was exchanged with media containing 5 µM JTE-013 + 5 µM S1P, while the contralateral S1P control eye received 5 µM S1P alone. Examples of outflow facility traces for experiments comparing S1P control (―) to JTE-013 + S1P experimental (…..) eyes are shown (A); paired eyes were from an 85 y/o donor (87C, 88E) with a net facility increase of 23.8% with the inclusion of JTE-013, compared to S1P control. Average normalized facilities following anterior chamber exchanges are shown (B). Data are presented as mean ± SD (* p<0.05).

Page 101: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

100 

 

5.4 Discussion

We originally hypothesized that the S1P1 receptor was responsible for the S1P-

mediated decrease in outflow facility due to receptor expression patterns in the outflow

pathway and predicted mechanisms of action, but surprisingly, activation of the S1P1

receptor alone did not have an effect on outflow facility, nor did it have an effect on MLC

phosphorylation in the cell culture models. Moreover, the use of a S1P1 or S1P1,3-

selective antagonist failed to block S1P-mediated MLC phosphorylation. Rather,

blocking the S1P2 receptor prevented S1P-promoted MLC phosphorylation in cultured

SC and TM cells. Thus, our focus shifted from the S1P1 to the S1P2 receptor, where

blocking S1P2 effectively prevented the entire S1P-mediated decrease in outflow facility

in human and porcine whole-eye perfusions. These results suggest that controlling S1P2

receptor activation in the conventional outflow pathway may provide a new

pharmacological target to reduce ocular hypertension in patients with, or are at risk for

POAG.

In this study, the effects observed with JTE-013 on outflow facility in both

porcine and human eyes demonstrates S1P2 to be a dominant receptor subtype in the

outflow tissue that may be responsible in mediating S1P’s effect of increasing outflow

resistance. JTE-013 blocked the S1P effect in human eyes despite not pretreating the

outflow tissue with antagonist as was done in the porcine experiments. The pretreatment

procedure was omitted due to donor age and length of time from TOD to start of

perfusion. We have found empirically that tissue responsiveness decreases with

increasing time between TOD and experimental treatment, as well as with tissue from

Page 102: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

101 

 

older donors. Despite the conservative approach, the human perfusion experiments

clearly demonstrate S1P2 to be the key receptor in the S1P-mediated effects in organ-

culture.

Three commercially available antagonists were tested to block S1P-promoted

MLC phosphorylation, a Rho/Rho-kinase-mediated effect observed robustly and reliably

in both SC and TM cells in this study [9, 151]. The S1P2 antagonist JTE-013

significantly reduced MLC phosphorylation by S1P in both SC and TM cell culture

models; in agreement with previous findings that S1P2 receptor activation increases MLC

phosphorylation in a Rho-dependent manner [111, 113]. JTE-013 is a selective S1P2

antagonist with IC50 values for human S1P1 and S1P2 receptors at >10 μM and ~17 nM,

respectively. For the S1P3 receptor, only 4.2% inhibition of S1P binding was observed at

10 μM JTE-013 [122]. However, recent evidence has suggested that JTE-013 may not be

highly selective to the S1P2 receptor at higher concentrations in rodents [154]. Therefore,

we used two different experimental setups in cell culture treatments using the S1P

receptor antagonists. First, we pretreated with a constant 10 μM antagonist

concentration, followed by S1P treatments of 10, 1, and 0.1 μM. Second, rather than

varying the S1P concentration, we varied the antagonist pretreatment concentrations (10,

1, and 0.1 μM) and followed with a constant 1 μM S1P treatment. Both experimental

setups demonstrated JTE-013 to block the S1P-promoted MLC phosphorylation in both

SC and TM cells, supporting S1P2 as the receptor involved in S1P-promoted MLC

phosphorylation.

Page 103: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

102 

 

The S1P2 receptor stimulates the Rho/Rho-kinase pathway, thus increasing MLC

phosphorylation by inhibiting MLC phosphatase [111, 113]. Activation of the Rho

pathway in the outflow tissue has been shown to increase outflow resistance [155] with

the contractile state of TM cells through MLC phosphorylation being the key regulating

event [9, 156]. Thus, S1P may promote increased TM cell contractility, thereby

constricting the passageways through the extracellular matrix in the JCT region of the

TM. Meanwhile, cell-cell and cell-matrix junctions in conjunction with a sustained

contractile state are vital to SC cells due to the large variation in trans-endothelial

pressure experienced by the inner wall SC cells. Thus, an S1P-mediated increase in Rho-

dependent MLC phosphorylation in SC cells at the cell cortex in conjunction with

cortical actin formation [151] may promote endothelial barrier enhancement with an

increase in cell-cell and cell-matrix connections, therefore raising outflow resistance.

These events in the TM and along the inner wall of SC, whether working individually or

in conjunction with one another, may prove to be the mechanism in the S1P-mediated

increase in outflow resistance.

Due to the exposure of the conventional outflow pathway to a wide range of flow

and pressure, the S1P receptor system may play a role in accommodating and responding

to changing conditions in the outflow tissue. Under normal conditions, basal activation

of the S1P receptor system may support normal cell-cell and cell-matrix interactions with

moderate actomyosin tone for both SC and TM cells. In the case of high outflow, the

spaces between TM cells and the extracellular matrix in the JCT are expanded, as well as

the sub-endothelial space of the inner wall. To help prevent detachment of SC at elevated

Page 104: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

103 

 

IOP, an increase in S1P release by outflow cells and subsequent saturation of the S1P

receptors may promote an increase in actomyosin tone via the S1P2/Rho/Rho-kinase

pathway to further increase stiffness in order to maintain the outflow structure. In

support of outflow cells increasing S1P release under high outflow conditions, vascular

cells have been shown to increase S1P release when under shear stress [157].

Accordingly, the shear stress experienced by the SC cells at elevated IOPs is predicted to

be comparable to the shear stress experienced by arterial vascular endothelial cells [129].

In conclusion, the S1P2 receptor may prove to be a valuable target to reduce

outflow resistance and IOP in glaucoma patients. S1P2 preferentially promotes a

contractile response in the cells of the outflow pathway, thus antagonizing S1P2 provides

a receptor-based target to increase aqueous outflow. In support of a potential S1P2-

specific antagonist to decrease outflow resistance in glaucoma patients, our perfusions

with JTE-013 alone displayed a trend towards increasing outflow facility in porcine eyes

while human eyes treated with JTE-013 + S1P displayed an increase of outflow facility

over baseline. Therefore, future studies using long-term perfusions with either JTE-013,

or another, more efficacious S1P2 antagonist are needed to verify S1P2 as a potential

glaucoma therapeutic.

Page 105: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

104 

 

6. CONCLUSIONS

6.1 Summary

My central hypothesis states that S1P receptor activation in Schlemm’s canal (SC)

contributes to decreased outflow facility of S1P-perfused human eyes in organ-culture.

To test this central hypothesis, I set out 3 specific aims. In specific aim 1, I characterized

the expression of S1P receptors in SC cells, originally hypothesizing the expression of the

S1P1 and S1P3 receptor subtypes due to their prominent expression in vascular

endothelial cells [11]. In chapter 3, I used immunoblot analysis, RT-PCR, and

immunofluorescence microscopy of frozen ocular sections to identify the S1P receptors

expressed. In the results, we found consistent and pronounced expression of the S1P1 and

S1P3 receptor subtypes, but limited expression of S1P2. In chapter 5, S1P2 –specific

antagonist JTE-013 (but not S1P1 or S1P3) blocked the S1P-induced MLC

phosphorylation in SC and trabecular meshwork (TM) primary cells, as well as the S1P-

induced decrease of outflow facility in the porcine perfusion model. The data from the

experiments using JTE-013 functionally support the expression of S1P2 in the

conventional outflow pathway.

For specific aim 2, I identified downstream effectors of S1P receptor activation in

SC cells. My original hypothesis, S1P-induced increase in actomyosin organization at

the cell cortex and promotion of junctional organization in SC cells, was based on S1P’s

known effect of decreasing endothelial permeability following an increase in junctional

organization [13, 104]. In chapter 4, I found that S1P activated both the Rac and Rho

GTPases, modulators of cytoskeletal dynamics, in SC cells. Also, S1P promoted myosin

Page 106: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

105 

 

light chain (MLC) phosphorylation in a time and dose-dependent manner similar to TM

cells, although not as robustly. The phospho-MLC species following S1P treatment were

localized at the cell cortex, similar to the cortical actin arrangement that was also

observed. In chapter 5, inclusion of an S1P2 antagonist blocked MLC phosphorylation by

S1P in both SC and TM cells, indicating that activation of S1P2 with the subsequent

promotion of a contractile state for cells in the conventional outflow pathway may be the

S1P mechanism in decreasing outflow facility. Although an increase in cortical

actomyosin arrangement was observed in S1P-treated SC cells, I could not determine if

S1P increases junctional formation due to the immature cell-cell junctions formed by SC

cells in culture.

For specific aim 3, I identified an S1P receptor responsible for S1P’s effect in the

whole-eye perfusion model. My original hypothesis, activation of the S1P1 receptor in

the conventional outflow pathway contributes to the S1P-induced decrease in outflow

facility, was based on the decrease of endothelial permeability following the activation of

S1P1 receptors in endothelial cells [13, 104] and the robust expression of S1P1 in SC cells

(chapter 3). In chapter 5, I perfused S1P receptor antagonists in both porcine and human

whole-eyes. In porcine eyes, the S1P-induced decrease of outflow facility was blocked

by the S1P2 antagonist JTE-013. Human eye perfusions displayed a trend towards JTE-

013 blocking the S1P effect. These results were in agreement to those found in the cell

culture models with MLC phosphorylation.

Page 107: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

106 

 

6.2 Interpretation

The identification of S1P receptor subtypes in the conventional outflow pathway

indicates that the S1P decrease of outflow facility in the whole-eye perfusion model is a

result of S1P receptor-mediated effects in SC and/or TM cells. Of the various

downstream effectors of S1P receptor activation, I specifically focused on investigating

the effectors affecting the actomyosin network due to the importance of cytoskeletal

dynamics in regulating outflow resistance. Additionally, the relatively quick response

with a significant and sustained decrease in outflow facility by S1P alluded to a change in

flow patterns with a decrease in routes for aqueous humor outflow, effects most likely

requiring cytoskeletal rearrangement.

S1P introduced to TM cells in the JCT preferentially increases stress fiber

formation and cell contractility, thus tightening the space between neighboring TM cells

and the underlying SC endothelia. While the TM cells themselves may not significantly

alter the flow of aqueous humor when they contract, the rearrangement of the loose

extracellular matrix and spaces surrounding the TM cells would. Meanwhile, in SC cells

of the inner wall, S1P preferentially increases cortical actomyosin formation that

potentially promotes an increase in endothelial barrier formation. It is important for SC

cells on the inner wall to have structural support with strong connections to neighboring

cells and the underlying basement membrane due to the wide range of pressure and flow

in the basal to apical direction that the inner wall encounters. Indeed, S1P promotion of

endothelial barrier formation includes an increase in the localization of focal adhesion

kinase, a key protein involved in integrin and cytoskeleton interaction, to cortical actin

Page 108: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

107 

 

formation [18]. A subsequent study has also shown interaction of focal adhesion kinase

with the adherens junction protein VE-cadherin through the adaptor protein β-catenin,

further supporting that S1P increases both cell-cell and cell-matrix interactions with

cortical actomyosin formation [106].

While the TM and inner wall of SC are separate layers of tissue, it is important to

view them as a single, functional unit in generating and regulating outflow resistance.

Along with forming cell-cell junctions to neighboring endothelial cells on the inner wall

and cell-matrix junctions to the underlying basement membrane, SC cells also form

contacts with TM cells in the juxtacanalicular connective tissue (JCT) region. These cell-

cell junctions through cytoplasmic processes are thought to help secure the inner wall

endothelia to its basal connections and prevent SC from collapsing in the face of large

trans-endothelial pressures [158]. The subsequent formation of giant vacuoles thus

allows the inner wall to withstand and respond to a wide-range of pressure encountered

under normal physiological conditions. When S1P is introduced to the conventional

outflow pathway, the heterotypic intercellular connections between SC and TM cells may

potentially be strengthened. The strengthening of these junctions, in addition to the

contractile state of the cells, would increase the resistance to outflow by further

compacting the space between the inner wall and the TM cells in the JCT.

Along with understanding how S1P may affect conventional outflow physiology,

it is also important to understand why the S1P receptor system is present in the outflow

pathway to begin with. We have observed prominent expression of both the S1P1 and

S1P3 receptors in cells of the outflow pathway, yet S1P2 was primarily responsible for the

Page 109: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

108 

 

S1P effect on outflow facility in the whole-eye perfusion model. One possible

explanation is that under basal flow and pressure, the S1P1 (Rac activation) and S1P3

(Rac and Rho activation) receptors may provide a role in maintaining normal cell-cell

and cell-matrix interactions. Activation of these receptors would provide actomyosin

tone to support the cell junctions, as well as generate moderate resistance to aqueous

outflow. When the S1P receptor system is saturated, such as during the whole-eye

perfusions with 5 μM S1P, a shift towards greater Rho signaling through the S1P2 (Rho

activation, Rac inactivation) and S1P3 receptors, resulting in increased cell contractility

may follow. A possible scenario in which saturating the S1P receptor system may come

into play is during high outflow. In the pressure-dependent conventional outflow

pathway, flow increases linearly with pressure. In a situation with high pressure and

flow, giant vacuoles form and spaces between neighboring TM cells, the extracellular

matrix, and the inner wall expands, thus narrowing the SC lumen. To prevent SC from

collapsing, increased S1P release from outflow cells may occur to increase S1P signaling

to maintain the outflow structure. In support of this idea, vascular endothelial cells have

been shown to release S1P when under shear stress [157]. Hence, S1P released from

outflow cells under heavy flow/pressure conditions may promote autocrine/paracrine S1P

signaling. The increase in Rho signaling through S1P2 and S1P3 may then counteract the

outflow forces.

Page 110: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

109 

 

6.3 Future direction

6.3.1 Sphingosine-1-phosphate perfusion

S1P promotes an immediate and significant drop of outflow facility within 20 -30

min following the introduction of S1P in both porcine and human eyes. While an acute

change in outflow facility was observed using an S1P concentration of 5μM, would long-

term perfusions at lower concentrations of S1P also decrease outflow facility to a similar

extent? To accomplish this, a long-term perfusion using the anterior segment perfusion

model would be better-suited over the acute whole-eye perfusion. In addition to long-

term perfusion with S1P, it would also be interesting to see whether inclusion of S1P

receptor antagonists alone would increase baseline outflow facility. This may give us

insight into whether there is basal S1P receptor activation with normal aqueous flow and

whether blocking that basal activation would increase outflow. If the S1P receptor

system is to become a novel target in the conventional outflow pathway to reduce ocular

hypertension, a S1P receptor antagonist-induced increase in facility would need to be

demonstrated. An example of an antagonist blocking basal S1P receptor activity levels

has been shown with the S1P1 antagonist W146 in increasing capillary leakage [121].

Another direction in investigating the role of the S1P system in regulating outflow

physiology is utilizing the mouse perfusion model. With the identification of S1P2 as a

key receptor in mediating S1P’s effect in both human and porcine eyes, we can now look

deeper into the receptor’s role using the S1p2-knockout mouse. Unlike the S1p1-knockout

mouse that is embryonic lethal [84], the S1p2-knockout animal is viable. Thus, we can

use the mouse eye perfusion to determine if S1P decreases outflow facility in the mouse

Page 111: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

110 

 

and whether that decrease is dependent on the S1P2 receptor. In addition, due to mice

having a similar SC to humans without as developed of a TM, we could assess the

contribution of the inner wall of SC to S1P’s effect on facility.

While S1P has been detected in aqueous humor [125], it is also important to

determine the source of S1P. The sphingosine kinases (SK) 1/2 are responsible for the

formation of S1P by phosphorylating sphingosine (Chapter 2.4.1). Future experiments

may include the localization of SK1/2 in the anterior segment of the eye. It would be

interesting to see whether SK is robustly expressed by TM cells, both on the trabecular

beams and the JCT, as this would be an ideal location to increase/decrease S1P levels

downstream to fellow TM or inner wall cells in order to regulate outflow resistance.

Thus, if SK is expressed in the outflow cells, would changes in pressure and/or flow

across TM cells grown on filters affect SK activity? Interestingly, vascular endothelial

cells under shear stress have been shown to increase S1P release [157]. Subsequent

perfusions with SK inhibitors may determine whether basal formation of S1P is required

to regulate a constant outflow resistance. A related experiment includes the perfusion of

a monoclonal S1P antibody (sonepcizumab) to bind endogenous S1P within the outflow

pathway. The S1P antibody has previously been shown to prevent S1P-promoted retinal

neovascularization [159].

In future perfusion whole-eye or anterior segment perfusion experiments,

inclusion of S1P may prove to be a valuable tool in detecting tissue responsiveness. As it

is, no experimental compound has been shown to rapidly reduce outflow facility, besides

S1P. Using S1P to test tissue responsiveness would be especially valuable in perfusion

Page 112: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

111 

 

experiments involving non-human eyes that undergo a volume-dependent increase in

outflow (washout effect) [130, 131, 160-162]. Most compounds of use in testing tissue

responsiveness increase outflow facility, a similar effect that occurs with washout.

Hence, S1P would be ideal due to its relatively quick (20-30 min) and significant

decrease in outflow facility. Other compounds that I have tried to perfuse for tissue

responsiveness have included cytochalasin-D, ATP, pilocarpine, and Y-27632.

Cytochalasin-D proved to be the most effective compound with a dramatic increase in

outflow facility at the end of the perfusion experiments, but it is a very potent compound

that disrupts the cytoskeleton wherever it comes in contact. Once again, S1P would be

better suited to test tissue responsiveness because its effects on the cytoskeleton are

receptor-mediated.

6.3.2 SC cell model

There is a significant drawback when using a cell culture model in research and

translating those findings to those particular cells in vivo. There is a loss of the native

conditions for those cells, such as the three-dimensional structural interactions to

neighboring cells and extracellular matrix and the presence of appropriate nutrients and

signaling molecules (paracrine and endocrine).

SC cells at confluence do not form junctions in a similar fashion that is observed

in vivo and perhaps new culturing techniques are needed to promote junction formation.

Proper cell-cell and cell-matrix junctions are required for hepatocytes in the liver for

normal functioning. Similar to SC cells, primary cultures of hepatocytes undergo a loss

Page 113: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

112 

 

of their cell junctions. Different strategies are employed to restore those junctions, such

as: 1) medium enrichment of monolayer cultures (e.g. corticosteroids, DMSO, cAMP), 2)

restoration of cell-cell contacts in co-cultures (e.g. co-culture with hepatic and non-

hepatic cells to restore intercellular contacts), and 3) reestablish cell-matrix contacts (e.g.

natural or synthetic extracellular matrix scaffold in a single, sandwich, or immobilization

culture) [163]. Similar strategies to restore cell junctions may be required to truly assess,

or at least gain a better understanding, of SC cell functionality in culture.

Page 114: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

113 

 

6.4 Perspective

Glaucoma, primary open-angle glaucoma in particular, is a disease of the aged.

As the average lifespan for people in developed countries increases, so too does the

prevalence for age-related diseases. While glaucoma is not a life-threatening ailment (at

least directly), the impact on the quality of life for patients living with glaucoma, as well

as the economic strain on care can be quite significant.

While current glaucoma therapeutics does reduce IOP effectively, glaucoma

patients usually become non-responsive to the drugs over time and often require multiple

drugs to control their IOP. The mechanisms for current glaucoma drugs to reduce IOP

include decreasing aqueous humor secretion, or increasing uveoscleral outflow. There is

no glaucoma drug on the market specifically aimed at the primary site for controlling

outflow resistance, the conventional outflow pathway. A drug targeting the

conventional outflow pathway, may not only prove to be more efficacious than current

options, but may also be the only drug needed throughout the patient’s lifetime to

maintain their IOP.

In conclusion, the data presented within this dissertation support the S1P receptor

system as a potential target within the conventional outflow pathway to regulate outflow

resistance and IOP. It will still take more research to determine whether a S1P receptor

antagonist would be considered clinically applicable to decrease elevated IOP.

Nevertheless, investigating the S1P receptor system has further increased our general

understanding as to how outflow resistance is regulated.

Page 115: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

114 

 

APPENDIX A: WHOLE-EYE PERFUSION MODEL

The whole-eye perfusion model used is based on the system described by Ethier

et. al. [128]. This system is commonly used for short-term experiments (6-8 hrs) to

observe acute effects during experimental ocular perfusions. The advantage of this

model is that it maintains the proper anatomy of the conventional outflow pathway in

vivo.

The perfusion program was developed in LabVIEW by Dr. Darryl Overby. The

pressure within the eye is monitored and recorded by a pressure transducer (Honeywell)

as a voltage reading. The voltage recorded by the pressure transducer is sent to the

computer (Dell) through a PCI-6221 data acquisition card (National Instruments). The

LabVIEW 7.1 software (National Instruments) then converts the voltage into IOP

(readings at different pressures were calibrated prior to each experiment). To maintain a

constant pressure perfusion, the flow rate from a syringe pump (Harvard Apparatus) is

controlled by the computer and altered to maintain the constant pressure in the eye. The

flow rate is modified by the following equation:

Qnew = Qold + Kp (Pold – Pnew) + Ki dt [Pset – (Pnew + Pold) /2]

where Kp = Q/P and Ki is Q/mmHg/dt.

The pressure tubing lines leading from the glass syringes (Hamilton) on the

syringe pumps (Harvard Apparatus) to the eyes and pressure transducer (Honeywell)

were filled with Dulbecco’s phosphate-buffered saline containing 5 mM glucose (DBG).

Prior to filling the lines, the DBG was de-gassed and sterile-filtered through 0.22 μm

syringe filter unit.

Page 116: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

115 

 

Enucleated whole-eyes were stored in a moist chamber with saline solution at 4ºC

following enucleation and prior to perfusion. Eyes were placed in a glass beaker

containing glass beads and gauze sponges to provide filling, structure, and padding for

the eyes. The glass beakers were filled to the top with phosphate-buffered saline, pH 7.4,

and placed in a water bath. The temperature of the water bath was set so that the

temperature of the saline in the glass beaker was maintained at 34ºC. The eyes were

placed in the beakers so that the saline level was at the limbal region of the eye (region

where cornea and sclera meet) and moist tissue was placed over the eyes to prevent the

cornea from drying out.

A 23 G infusion needle connecting the rest of the lines (syringe pump, pressure

transducer, etc.) to the eye was inserted through the pupil and into the posterior chamber

of the eye beneath the iris. Care was taken to ensure the beveled-side

(opening) of the needle was not pressed against either the iris or the lens. The lines

leading to the eye were then opened up to a pressure head set at either 8 mmHg (human)

or 15 mmHg (porcine) and IOP was allowed to establish for 15 min. At the end of the 15

min acclimating period, constant pressure perfusion at either 8 mmHg (human) or 15

mmHg (porcine) began.

Supplies required for whole-eye perfusion:

− Hamilton gastight syringe, 2.5 ml, 22 G (81420)

− Becton Dickinson vacutainer blood collection set, 23G x 3/4” x 12” (367253)

− Becton Dickinson 10 ml sterile plastic syringe (309604)

Page 117: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

116 

 

− Becton Dickinson 20 ml sterile plastic syringe (309661)

− Becton Dickinson Falcon sterile petri dish, 100 x 15 mm (351029)

− Kendall Argyle Ez-Flo 3-way stopcock (173518)

− Mallinkrodt pressure tubing, 0.05” x 36” (91113)

− Gauze sponges

− Kimwipes

− Millipore Millex syringe driven filter unit, 0.22 μm pore, PES membrane

− Dulbecco’s phosphate-buffered saline, with calcium and magnesium

− (D)-glucose

Equipment required for whole-eye perfusion:

− Honeywell ultra-low pressure sensor (163PC01D48)

− Omega Engineering adjustable power supply (PST-4130)

− Harvard Apparatus PHD 22/2000 infusion only syringe pump (70-2000)

− Harvard Apparatus Daisy-chain connector, 9 pin D-sub (702022)

− Harvard Apparatus Daisy-chain cable, 1.8 m (722478)

− Fisher Scientific Isotemp general purpose water bath, 2 L shallow (202S)

Data acquisition and analysis:

− Dell Dimension 3000 computer with monitor

− National Instruments Data acquisition (DAQ) card for Windows (PCI-6221)

− National Instruments LabVIEW 7.1 for Windows

− Microsoft Office Excel 2007

Page 118: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

117 

 

REFERENCES

1. Quigley, H.A. and A.T. Broman, The number of people with glaucoma worldwide in 2010 and 2020. Br J Ophthalmol, 2006. 90(3): p. 262-7.

2. Foster, P.J., et al., The definition and classification of glaucoma in prevalence

surveys. Br J Ophthalmol, 2002. 86(2): p. 238-42. 3. Quigley, H.A. and W.R. Green, The histology of human glaucoma cupping and

optic nerve damage: clinicopathologic correlation in 21 eyes. Ophthalmology, 1979. 86(10): p. 1803-30.

4. Quigley, H.A., et al., Optic nerve damage in human glaucoma. II. The site of

injury and susceptibility to damage. Arch Ophthalmol, 1981. 99(4): p. 635-49. 5. Grant, W.M., Experimental aqueous perfusion in enucleated human eyes. Arch

Ophthalmol, 1963. 69: p. 783-801. 6. Rosenquist, R., et al., Outflow resistance of enucleated human eyes at two

different perfusion pressures and different extents of trabeculotomy. Curr Eye Res, 1989. 8(12): p. 1233-40.

7. Maepea, O. and A. Bill, Pressures in the juxtacanalicular tissue and Schlemm's

canal in monkeys. Exp Eye Res, 1992. 54(6): p. 879-83. 8. Johnson, M., et al., Modulation of outflow resistance by the pores of the inner

wall endothelium. Invest Ophthalmol Vis Sci, 1992. 33(5): p. 1670-5. 9. Mettu, P.S., et al., Role of lysophospholipid growth factors in the modulation of

aqueous humor outflow facility. Invest Ophthalmol Vis Sci, 2004. 45(7): p. 2263-71.

10. Lee, M.J., et al., Sphingosine-1-phosphate as a ligand for the G protein-coupled

receptor EDG-1. Science, 1998. 279(5356): p. 1552-5. 11. Takuwa, Y., et al., Sphingosine-1-phosphate signaling and biological activities in

the cardiovascular system. Biochim Biophys Acta, 2008. 1781(9): p. 483-8. 12. Rosen, H., et al., Sphingosine 1-phosphate receptor signaling. Annu Rev

Biochem, 2009. 78: p. 743-68.

Page 119: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

118 

 

13. Donati, C. and P. Bruni, Sphingosine 1-phosphate regulates cytoskeleton dynamics: implications in its biological response. Biochim Biophys Acta, 2006. 1758(12): p. 2037-48.

14. Mehta, D., et al., Sphingosine 1-phosphate-induced mobilization of intracellular

Ca2+ mediates rac activation and adherens junction assembly in endothelial cells. J Biol Chem, 2005. 280(17): p. 17320-8.

15. Lee, M.J., et al., Vascular endothelial cell adherens junction assembly and

morphogenesis induced by sphingosine-1-phosphate. Cell, 1999. 99(3): p. 301-12. 16. Lee, J.F., et al., Dual roles of tight junction-associated protein, zonula occludens-

1, in sphingosine 1-phosphate-mediated endothelial chemotaxis and barrier integrity. J Biol Chem, 2006. 281(39): p. 29190-200.

17. Garcia, J.G., et al., Sphingosine 1-phosphate promotes endothelial cell barrier

integrity by Edg-dependent cytoskeletal rearrangement. J Clin Invest, 2001. 108(5): p. 689-701.

18. Shikata, Y., K.G. Birukov, and J.G. Garcia, S1P induces FA remodeling in human

pulmonary endothelial cells: role of Rac, GIT1, FAK, and paxillin. J Appl Physiol, 2003. 94(3): p. 1193-203.

19. Dudek, S.M., et al., Pulmonary endothelial cell barrier enhancement by

sphingosine 1-phosphate: roles for cortactin and myosin light chain kinase. J Biol Chem, 2004. 279(23): p. 24692-700.

20. Ramos, R.F., et al., Schlemm's canal endothelia, lymphatic, or blood vasculature?

J Glaucoma, 2007. 16(4): p. 391-405. 21. Quigley, H.A., Open-angle glaucoma. N Engl J Med, 1993. 328(15): p. 1097-106. 22. Quigley, H.A., Number of people with glaucoma worldwide. Br J Ophthalmol,

1996. 80(5): p. 389-93. 23. Tielsch, J.M., et al., Racial variations in the prevalence of primary open-angle

glaucoma. The Baltimore Eye Survey. JAMA, 1991. 266(3): p. 369-74. 24. Quigley, H.A. and S. Vitale, Models of open-angle glaucoma prevalence and

incidence in the United States. Invest Ophthalmol Vis Sci, 1997. 38(1): p. 83-91. 25. Wolfs, R.C., et al., Genetic risk of primary open-angle glaucoma. Population-

based familial aggregation study. Arch Ophthalmol, 1998. 116(12): p. 1640-5.

Page 120: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

119 

 

26. Wiggs, J.L., Genetic etiologies of glaucoma. Arch Ophthalmol, 2007. 125(1): p.

30-7. 27. Nemesure, B., et al., Incident open-angle glaucoma and intraocular pressure.

Ophthalmology, 2007. 114(10): p. 1810-5. 28. Sommer, A., et al., Relationship between intraocular pressure and primary open

angle glaucoma among white and black Americans. The Baltimore Eye Survey. Arch Ophthalmol, 1991. 109(8): p. 1090-5.

29. Leske, M.C., Open-angle glaucoma -- an epidemiologic overview. Ophthalmic

Epidemiol, 2007. 14(4): p. 166-72. 30. Anderson, D.R. and A. Hendrickson, Effect of intraocular pressure on rapid

axoplasmic transport in monkey optic nerve. Invest Ophthalmol, 1974. 13(10): p. 771-83.

31. Minckler, D.S., A.H. Bunt, and G.W. Johanson, Orthograde and retrograde

axoplasmic transport during acute ocular hypertension in the monkey. Invest Ophthalmol Vis Sci, 1977. 16(5): p. 426-41.

32. Quigley, H. and D.R. Anderson, The dynamics and location of axonal transport

blockade by acute intraocular pressure elevation in primate optic nerve. Invest Ophthalmol, 1976. 15(8): p. 606-16.

33. Howell, G.R., et al., Axons of retinal ganglion cells are insulted in the optic nerve

early in DBA/2J glaucoma. J Cell Biol, 2007. 179(7): p. 1523-37. 34. The Advanced Glaucoma Intervention Study (AGIS): 7. The relationship between

control of intraocular pressure and visual field deterioration.The AGIS Investigators. Am J Ophthalmol, 2000. 130(4): p. 429-40.

35. Ritch, R., M.B. Shields, and T. Krupin, The glaucomas. 2nd ed. 1996, St. Louis:

Mosby. 3 v. (xxvi, 1807, 27 p.). 36. Civan, M.M. and A.D. Macknight, The ins and outs of aqueous humour secretion.

Exp Eye Res, 2004. 78(3): p. 625-31. 37. Alm, A. and S.F. Nilsson, Uveoscleral outflow--a review. Exp Eye Res, 2009.

88(4): p. 760-8.

Page 121: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

120 

 

38. Bill, A. and C.I. Phillips, Uveoscleral drainage of aqueous humour in human eyes. Exp Eye Res, 1971. 12(3): p. 275-81.

39. Bill, A., Conventional and uveo-scleral drainage of aqueous humour in the

cynomolgus monkey (Macaca irus) at normal and high intraocular pressures. Exp Eye Res, 1966. 5(1): p. 45-54.

40. Gabelt, B.T., et al., Aqueous humor dynamics and trabecular meshwork and

anterior ciliary muscle morphologic changes with age in rhesus monkeys. Invest Ophthalmol Vis Sci, 2003. 44(5): p. 2118-25.

41. Maepea, O. and A. Bill, The pressures in the episcleral veins, Schlemm's canal

and the trabecular meshwork in monkeys: effects of changes in intraocular pressure. Exp Eye Res, 1989. 49(4): p. 645-63.

42. Johnson, M., 'What controls aqueous humour outflow resistance?'. Exp Eye Res,

2006. 82(4): p. 545-57. 43. Gong, H., R.C. Tripathi, and B.J. Tripathi, Morphology of the aqueous outflow

pathway. Microsc Res Tech, 1996. 33(4): p. 336-67. 44. Epstein, D.L., et al., Experimental obstruction to aqueous outflow by pigment

particles in living monkeys. Invest Ophthalmol Vis Sci, 1986. 27(3): p. 387-95. 45. Richardson, T.M., B.T. Hutchinson, and W.M. Grant, The outflow tract in

pigmentary glaucoma: a light and electron microscopic study. Arch Ophthalmol, 1977. 95(6): p. 1015-25.

46. Buller, C., D.H. Johnson, and R.C. Tschumper, Human trabecular meshwork

phagocytosis. Observations in an organ culture system. Invest Ophthalmol Vis Sci, 1990. 31(10): p. 2156-63.

47. Alvarado, J., et al., Age-related changes in trabecular meshwork cellularity.

Invest Ophthalmol Vis Sci, 1981. 21(5): p. 714-27. 48. de Kater, A.W., S. Melamed, and D.L. Epstein, Patterns of aqueous humor

outflow in glaucomatous and nonglaucomatous human eyes. A tracer study using cationized ferritin. Arch Ophthalmol, 1989. 107(4): p. 572-6.

49. Tripathi, R.C., Ultrastructure of Schlemm's canal in relation to aqueous outflow.

Exp Eye Res, 1968. 7(3): p. 335-41.

Page 122: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

121 

 

50. Ye, W., et al., Interendothelial junctions in normal human Schlemm's canal respond to changes in pressure. Invest Ophthalmol Vis Sci, 1997. 38(12): p. 2460-8.

51. Ethier, C.R., et al., Two pore types in the inner-wall endothelium of Schlemm's

canal. Invest Ophthalmol Vis Sci, 1998. 39(11): p. 2041-8. 52. Allingham, R.R., et al., The relationship between pore density and outflow facility

in human eyes. Invest Ophthalmol Vis Sci, 1992. 33(5): p. 1661-9. 53. Sit, A.J., et al., Factors affecting the pores of the inner wall endothelium of

Schlemm's canal. Invest Ophthalmol Vis Sci, 1997. 38(8): p. 1517-25. 54. Ethier, C.R., The inner wall of Schlemm's canal. Exp Eye Res, 2002. 74(2): p.

161-72. 55. Garron, L.K., et al., Electron microscopic studies of the human eye. I. Preliminary

investigations of the trabeculas. Am J Ophthalmol, 1958. 46(1, Part 2): p. 27-35. 56. Tripathi, R.C., Mechanism of the aqueous outflow across the trabecular wall of

Schlemm's canal. Exp Eye Res, 1971. 11(1): p. 116-21. 57. Johnstone, M.A. and W.G. Grant, Pressure-dependent changes in structures of

the aqueous outflow system of human and monkey eyes. Am J Ophthalmol, 1973. 75(3): p. 365-83.

58. Brilakis, H.S. and D.H. Johnson, Giant vacuole survival time and implications for

aqueous humor outflow. J Glaucoma, 2001. 10(4): p. 277-83. 59. Parc, C.E., D.H. Johnson, and H.S. Brilakis, Giant vacuoles are found

preferentially near collector channels. Invest Ophthalmol Vis Sci, 2000. 41(10): p. 2984-90.

60. Inomata, H., A. Bill, and G.K. Smelser, Aqueous humor pathways through the

trabecular meshwork and into Schlemm's canal in the cynomolgus monkey (Macaca irus). An electron microscopic study. Am J Ophthalmol, 1972. 73(5): p. 760-89.

61. Tian, B., et al., The role of the actomyosin system in regulating trabecular fluid

outflow. Exp Eye Res, 2009. 88(4): p. 713-7.

Page 123: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

122 

 

62. Ethier, C.R., A.T. Read, and D.W. Chan, Effects of latrunculin-B on outflow facility and trabecular meshwork structure in human eyes. Invest Ophthalmol Vis Sci, 2006. 47(5): p. 1991-8.

63. Kaufman, P.L. and K.A. Erickson, Cytochalasin B and D dose-outflow facility

response relationships in the cynomolgus monkey. Invest Ophthalmol Vis Sci, 1982. 23(5): p. 646-50.

64. Johnson, D.H., The effect of cytochalasin D on outflow facility and the trabecular

meshwork of the human eye in perfusion organ culture. Invest Ophthalmol Vis Sci, 1997. 38(13): p. 2790-9.

65. Honjo, M., et al., Effects of rho-associated protein kinase inhibitor Y-27632 on

intraocular pressure and outflow facility. Invest Ophthalmol Vis Sci, 2001. 42(1): p. 137-44.

66. Rao, P.V., et al., Modulation of aqueous humor outflow facility by the Rho kinase-

specific inhibitor Y-27632. Invest Ophthalmol Vis Sci, 2001. 42(5): p. 1029-37. 67. Lu, Z., et al., The mechanism of increasing outflow facility by rho-kinase

inhibition with Y-27632 in bovine eyes. Exp Eye Res, 2008. 86(2): p. 271-81. 68. Tian, B., L.C. Brumback, and P.L. Kaufman, ML-7, chelerythrine and phorbol

ester increase outflow facility in the monkey Eye. Exp Eye Res, 2000. 71(6): p. 551-66.

69. Murata, N., et al., Quantitative measurement of sphingosine 1-phosphate by

radioreceptor-binding assay. Anal Biochem, 2000. 282(1): p. 115-20. 70. Murata, N., et al., Interaction of sphingosine 1-phosphate with plasma

components, including lipoproteins, regulates the lipid receptor-mediated actions. Biochem J, 2000. 352 Pt 3: p. 809-15.

71. Hla, T. and T. Maciag, An abundant transcript induced in differentiating human

endothelial cells encodes a polypeptide with structural similarities to G-protein-coupled receptors. J Biol Chem, 1990. 265(16): p. 9308-13.

72. Le Stunff, H., S. Milstien, and S. Spiegel, Generation and metabolism of

bioactive sphingosine-1-phosphate. J Cell Biochem, 2004. 92(5): p. 882-99. 73. Xia, P., et al., Tumor necrosis factor-alpha induces adhesion molecule expression

through the sphingosine kinase pathway. Proc Natl Acad Sci U S A, 1998. 95(24): p. 14196-201.

Page 124: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

123 

 

74. Pettus, B.J., et al., The sphingosine kinase 1/sphingosine-1-phosphate pathway

mediates COX-2 induction and PGE2 production in response to TNF-alpha. FASEB J, 2003. 17(11): p. 1411-21.

75. Ancellin, N., et al., Extracellular export of sphingosine kinase-1 enzyme.

Sphingosine 1-phosphate generation and the induction of angiogenic vascular maturation. J Biol Chem, 2002. 277(8): p. 6667-75.

76. Mitra, P., et al., Role of ABCC1 in export of sphingosine-1-phosphate from mast

cells. Proc Natl Acad Sci U S A, 2006. 103(44): p. 16394-9. 77. Kobayashi, N., A. Yamaguchi, and T. Nishi, Characterization of the ATP-

dependent sphingosine 1-phosphate transporter in rat erythrocytes. J Biol Chem, 2009. 284(32): p. 21192-200.

78. Kobayashi, N., et al., Sphingosine 1-phosphate is released from the cytosol of rat

platelets in a carrier-mediated manner. J Lipid Res, 2006. 47(3): p. 614-21. 79. Sato, K., et al., Critical role of ABCA1 transporter in sphingosine 1-phosphate

release from astrocytes. J Neurochem, 2007. 80. Okamoto, H., et al., EDG1 is a functional sphingosine-1-phosphate receptor that

is linked via a Gi/o to multiple signaling pathways, including phospholipase C activation, Ca2+ mobilization, Ras-mitogen-activated protein kinase activation, and adenylate cyclase inhibition. J Biol Chem, 1998. 273(42): p. 27104-10.

81. Chun, J., et al., International Union of Pharmacology. XXXIV. Lysophospholipid

receptor nomenclature. Pharmacol Rev, 2002. 54(2): p. 265-9. 82. Ancellin, N. and T. Hla, Differential pharmacological properties and signal

transduction of the sphingosine 1-phosphate receptors EDG-1, EDG-3, and EDG-5. J Biol Chem, 1999. 274(27): p. 18997-9002.

83. Windh, R.T., et al., Differential coupling of the sphingosine 1-phosphate

receptors Edg-1, Edg-3, and H218/Edg-5 to the G(i), G(q), and G(12) families of heterotrimeric G proteins. J Biol Chem, 1999. 274(39): p. 27351-8.

84. Liu, Y., et al., Edg-1, the G protein-coupled receptor for sphingosine-1-

phosphate, is essential for vascular maturation. J Clin Invest, 2000. 106(8): p. 951-61.

Page 125: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

124 

 

85. Paik, J.H., et al., Sphingosine 1-phosphate receptor regulation of N-cadherin mediates vascular stabilization. Genes Dev, 2004. 18(19): p. 2392-403.

86. Krump-Konvalinkova, V., et al., Stable knock-down of the sphingosine 1-

phosphate receptor S1P1 influences multiple functions of human endothelial cells. Arterioscler Thromb Vasc Biol, 2005. 25(3): p. 546-52.

87. Ishii, I., et al., Selective loss of sphingosine 1-phosphate signaling with no obvious

phenotypic abnormality in mice lacking its G protein-coupled receptor, LP(B3)/EDG-3. J Biol Chem, 2001. 276(36): p. 33697-704.

88. MacLennan, A.J., et al., An essential role for the H218/AGR16/Edg-5/LP(B2)

sphingosine 1-phosphate receptor in neuronal excitability. Eur J Neurosci, 2001. 14(2): p. 203-9.

89. Kono, M., et al., Deafness and stria vascularis defects in S1P2 receptor-null mice.

J Biol Chem, 2007. 282(14): p. 10690-6. 90. Kono, M., et al., The sphingosine-1-phosphate receptors S1P1, S1P2, and S1P3

function coordinately during embryonic angiogenesis. J Biol Chem, 2004. 279(28): p. 29367-73.

91. Takuwa, Y., Subtype-specific differential regulation of Rho family G proteins and

cell migration by the Edg family sphingosine-1-phosphate receptors. Biochim Biophys Acta, 2002. 1582(1-3): p. 112-20.

92. Vetter, I.R. and A. Wittinghofer, The guanine nucleotide-binding switch in three

dimensions. Science, 2001. 294(5545): p. 1299-304. 93. Ridley, A.J., Rho GTPases and cell migration. J Cell Sci, 2001. 114(Pt 15): p.

2713-22. 94. English, D., et al., Induction of endothelial cell chemotaxis by sphingosine 1-

phosphate and stabilization of endothelial monolayer barrier function by lysophosphatidic acid, potential mediators of hematopoietic angiogenesis. J Hematother Stem Cell Res, 1999. 8(6): p. 627-34.

95. Wang, F., et al., Sphingosine 1-phosphate stimulates cell migration through a

G(i)-coupled cell surface receptor. Potential involvement in angiogenesis. J Biol Chem, 1999. 274(50): p. 35343-50.

Page 126: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

125 

 

96. Kimura, T., et al., Sphingosine 1-phosphate stimulates proliferation and migration of human endothelial cells possibly through the lipid receptors, Edg-1 and Edg-3. Biochem J, 2000. 348 Pt 1: p. 71-6.

97. Liu, F., et al., Differential regulation of sphingosine-1-phosphate- and VEGF-

induced endothelial cell chemotaxis. Involvement of G(ialpha2)-linked Rho kinase activity. Am J Respir Cell Mol Biol, 2001. 24(6): p. 711-9.

98. Sugimoto, N., et al., Inhibitory and stimulatory regulation of Rac and cell motility

by the G12/13-Rho and Gi pathways integrated downstream of a single G protein-coupled sphingosine-1-phosphate receptor isoform. Mol Cell Biol, 2003. 23(5): p. 1534-45.

99. Lepley, D., et al., The G protein-coupled receptor S1P2 regulates Rho/Rho kinase

pathway to inhibit tumor cell migration. Cancer Res, 2005. 65(9): p. 3788-95. 100. Takashima, S., et al., G12/13 and Gq mediate S1P2-induced inhibition of Rac and

migration in vascular smooth muscle in a manner dependent on Rho but not Rho kinase. Cardiovasc Res, 2008. 79(4): p. 689-97.

101. Hobson, J.P., et al., Role of the sphingosine-1-phosphate receptor EDG-1 in

PDGF-induced cell motility. Science, 2001. 291(5509): p. 1800-3. 102. Paik, J.H., et al., Sphingosine 1-phosphate-induced endothelial cell migration

requires the expression of EDG-1 and EDG-3 receptors and Rho-dependent activation of alpha vbeta3- and beta1-containing integrins. J Biol Chem, 2001. 276(15): p. 11830-7.

103. Wojciak-Stothard, B., et al., Rho and Rac but not Cdc42 regulate endothelial cell

permeability. J Cell Sci, 2001. 114(Pt 7): p. 1343-55. 104. McVerry, B.J. and J.G. Garcia, In vitro and in vivo modulation of vascular

barrier integrity by sphingosine 1-phosphate: mechanistic insights. Cell Signal, 2005. 17(2): p. 131-9.

105. Li, Y., et al., Interaction of cortactin and Arp2/3 complex is required for

sphingosine-1-phosphate-induced endothelial cell remodeling. Exp Cell Res, 2004. 298(1): p. 107-21.

106. Sun, X., et al., Enhanced interaction between focal adhesion and adherens

junction proteins: involvement in sphingosine 1-phosphate-induced endothelial barrier enhancement. Microvasc Res, 2009. 77(3): p. 304-13.

Page 127: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

126 

 

107. Singleton, P.A., et al., Regulation of sphingosine 1-phosphate-induced endothelial cytoskeletal rearrangement and barrier enhancement by S1P1 receptor, PI3 kinase, Tiam1/Rac1, and alpha-actinin. FASEB J, 2005. 19(12): p. 1646-56.

108. Goeckeler, Z.M. and R.B. Wysolmerski, Myosin light chain kinase-regulated

endothelial cell contraction: the relationship between isometric tension, actin polymerization, and myosin phosphorylation. J Cell Biol, 1995. 130(3): p. 613-27.

109. Garcia, J.G., H.W. Davis, and C.E. Patterson, Regulation of endothelial cell gap

formation and barrier dysfunction: role of myosin light chain phosphorylation. J Cell Physiol, 1995. 163(3): p. 510-22.

110. Vandenbroucke, E., et al., Regulation of endothelial junctional permeability. Ann

N Y Acad Sci, 2008. 1123: p. 134-45. 111. Gonda, K., et al., The novel sphingosine 1-phosphate receptor AGR16 is coupled

via pertussis toxin-sensitive and -insensitive G-proteins to multiple signalling pathways. Biochem J, 1999. 337 ( Pt 1): p. 67-75.

112. Okamoto, H., et al., Inhibitory regulation of Rac activation, membrane ruffling,

and cell migration by the G protein-coupled sphingosine-1-phosphate receptor EDG5 but not EDG1 or EDG3. Mol Cell Biol, 2000. 20(24): p. 9247-61.

113. Zhou, H. and K.S. Murthy, Distinctive G protein-dependent signaling in smooth

muscle by sphingosine 1-phosphate receptors S1P1 and S1P2. Am J Physiol Cell Physiol, 2004. 286(5): p. C1130-8.

114. Sanchez, T., et al., Induction of vascular permeability by the sphingosine-1-

phosphate receptor-2 (S1P2R) and its downstream effectors ROCK and PTEN. Arterioscler Thromb Vasc Biol, 2007. 27(6): p. 1312-8.

115. Skoura, A., et al., Essential role of sphingosine 1-phosphate receptor 2 in

pathological angiogenesis of the mouse retina. J Clin Invest, 2007. 117(9): p. 2506-16.

116. Marsolais, D. and H. Rosen, Chemical modulators of sphingosine-1-phosphate

receptors as barrier-oriented therapeutic molecules. Nat Rev Drug Discov, 2009. 8(4): p. 297-307.

117. Mandala, S., et al., Alteration of lymphocyte trafficking by sphingosine-1-

phosphate receptor agonists. Science, 2002. 296(5566): p. 346-9.

Page 128: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

127 

 

118. Graler, M.H. and E.J. Goetzl, The immunosuppressant FTY720 down-regulates sphingosine 1-phosphate G-protein-coupled receptors. FASEB J, 2004. 18(3): p. 551-3.

119. Kappos, L., et al., Oral fingolimod (FTY720) for relapsing multiple sclerosis. N

Engl J Med, 2006. 355(11): p. 1124-40. 120. Lien, Y.H., et al., S1P(1)-selective agonist, SEW2871, ameliorates ischemic acute

renal failure. Kidney Int, 2006. 69(9): p. 1601-8. 121. Sanna, M.G., et al., Enhancement of capillary leakage and restoration of

lymphocyte egress by a chiral S1P1 antagonist in vivo. Nat Chem Biol, 2006. 2(8): p. 434-41.

122. Osada, M., et al., Modulation of sphingosine 1-phosphate/EDG signaling by

tumor necrosis factor-alpha in vascular endothelial cells. Thromb Res, 2002. 108(2-3): p. 169-74.

123. Hamanaka, T., et al., Aspects of the development of Schlemm's canal. Exp Eye

Res, 1992. 55(3): p. 479-88. 124. Panetti, T.S., Differential effects of sphingosine 1-phosphate and lysophosphatidic

acid on endothelial cells. Biochim Biophys Acta, 2002. 1582(1-3): p. 190-6. 125. Liliom, K., et al., Growth factor-like phospholipids generated after corneal

injury. Am J Physiol, 1998. 274(4 Pt 1): p. C1065-74. 126. Stamer, W.D., et al., Isolation, culture, and characterization of endothelial cells

from Schlemm's canal. Invest Ophthalmol Vis Sci, 1998. 39(10): p. 1804-12. 127. Stamer, W.D., et al., Isolation and culture of human trabecular meshwork cells by

extracellular matrix digestion. Curr Eye Res, 1995. 14(7): p. 611-7. 128. Ethier, C.R., P. Ajersch, and R. Pirog, An improved ocular perfusion system. Curr

Eye Res, 1993. 12(8): p. 765-70. 129. Ethier, C.R., A.T. Read, and D. Chan, Biomechanics of Schlemm's canal

endothelial cells: influence on F-actin architecture. Biophys J, 2004. 87(4): p. 2828-37.

130. Overby, D., et al., The mechanism of increasing outflow facility during washout in

the bovine eye. Invest Ophthalmol Vis Sci, 2002. 43(11): p. 3455-64.

Page 129: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

128 

 

131. Scott, P.A., et al., Comparative studies between species that do and do not exhibit the washout effect. Exp Eye Res, 2007. 84(3): p. 435-43.

132. Tian, B., et al., H-7 disrupts the actin cytoskeleton and increases outflow facility.

Arch Ophthalmol, 1998. 116(5): p. 633-43. 133. Ethier, C.R. and D.W. Chan, Cationic ferritin changes outflow facility in human

eyes whereas anionic ferritin does not. Invest Ophthalmol Vis Sci, 2001. 42(8): p. 1795-802.

134. Sanchez, T., et al., Phosphorylation and action of the immunomodulator FTY720

inhibits vascular endothelial cell growth factor-induced vascular permeability. J Biol Chem, 2003. 278(47): p. 47281-90.

135. Vittet, D., et al., Targeted null-mutation in the vascular endothelial-cadherin gene

impairs the organization of vascular-like structures in embryoid bodies. Proc Natl Acad Sci U S A, 1997. 94(12): p. 6273-8.

136. Carmeliet, P., et al., Targeted deficiency or cytosolic truncation of the VE-

cadherin gene in mice impairs VEGF-mediated endothelial survival and angiogenesis. Cell, 1999. 98(2): p. 147-57.

137. Corada, M., et al., Vascular endothelial-cadherin is an important determinant of

microvascular integrity in vivo. Proc Natl Acad Sci U S A, 1999. 96(17): p. 9815-20.

138. Gumbiner, B.M., Regulation of cadherin adhesive activity. J Cell Biol, 2000.

148(3): p. 399-404. 139. Rajasekaran, A.K., et al., Catenins and zonula occludens-1 form a complex during

early stages in the assembly of tight junctions. J Cell Biol, 1996. 132(3): p. 451-63.

140. Roura, S., et al., Regulation of E-cadherin/Catenin association by tyrosine

phosphorylation. J Biol Chem, 1999. 274(51): p. 36734-40. 141. Wong, E.Y., et al., Vascular endothelial growth factor stimulates

dephosphorylation of the catenins p120 and p100 in endothelial cells. Biochem J, 2000. 346 Pt 1: p. 209-16.

142. Xia, X., D.J. Mariner, and A.B. Reynolds, Adhesion-associated and PKC-

modulated changes in serine/threonine phosphorylation of p120-catenin. Biochemistry, 2003. 42(30): p. 9195-204.

Page 130: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

129 

 

143. Chiu, Y.J., E. McBeath, and K. Fujiwara, Mechanotransduction in an extracted

cell model: Fyn drives stretch- and flow-elicited PECAM-1 phosphorylation. J Cell Biol, 2008. 182(4): p. 753-63.

144. Huang, Y.T., et al., Sphingosine 1-phosphate induces platelet/endothelial cell

adhesion molecule-1 phosphorylation in human endothelial cells through cSrc and Fyn. Cell Signal, 2008. 20(8): p. 1521-7.

145. Overby, D.R., W.D. Stamer, and M. Johnson, The changing paradigm of outflow

resistance generation: towards synergistic models of the JCT and inner wall endothelium. Exp Eye Res, 2009. 88(4): p. 656-70.

146. Wan, Z., D.F. Woodward, and W.D. Stamer, Endogenous Bioactive Lipids and

the Regulation of Conventional Outflow Facility. Expert Rev Ophthalmol, 2008. 3(4): p. 457-470.

147. Stamer, W.D., et al., Sphingosine-1-phosphate effects on the inner wall of

Schlemm's canal and outflow facility in perfused human eyes. Exp Eye Res, 2009. 89(6): p. 980-8.

148. Wang, L. and S.M. Dudek, Regulation of vascular permeability by sphingosine 1-

phosphate. Microvasc Res, 2009. 77(1): p. 39-45. 149. Ren, X.D., W.B. Kiosses, and M.A. Schwartz, Regulation of the small GTP-

binding protein Rho by cell adhesion and the cytoskeleton. EMBO J, 1999. 18(3): p. 578-85.

150. Kimura, K., et al., Regulation of myosin phosphatase by Rho and Rho-associated

kinase (Rho-kinase). Science, 1996. 273(5272): p. 245-8. 151. Sumida, G.M. and W.D. Stamer, Sphingosine-1-phosphate enhances cortical

actomyosin organization in cultured human Schlemm's canal endothelial cell monolayers. Invest Ophthalmol Vis Sci.

152. Sanna, M.G., et al., Sphingosine 1-phosphate (S1P) receptor subtypes S1P1 and

S1P3, respectively, regulate lymphocyte recirculation and heart rate. J Biol Chem, 2004. 279(14): p. 13839-48.

153. Davis, M.D., et al., Sphingosine 1-phosphate analogs as receptor antagonists. J

Biol Chem, 2005. 280(11): p. 9833-41.

Page 131: SPHINGOSINE-1-PHOSPHATE EFFECTS ON CONVENTIONAL ...

130 

 

154. Salomone, S., et al., Analysis of sphingosine 1-phosphate receptors involved in constriction of isolated cerebral arteries with receptor null mice and pharmacological tools. Br J Pharmacol, 2008. 153(1): p. 140-7.

155. Zhang, M., R. Maddala, and P.V. Rao, Novel molecular insights into RhoA

GTPase-induced resistance to aqueous humor outflow through the trabecular meshwork. Am J Physiol Cell Physiol, 2008. 295(5): p. C1057-70.

156. Rao, P.V., et al., Regulation of myosin light chain phosphorylation in the

trabecular meshwork: role in aqueous humour outflow facility. Exp Eye Res, 2005. 80(2): p. 197-206.

157. Venkataraman, K., et al., Vascular endothelium as a contributor of plasma

sphingosine 1-phosphate. Circ Res, 2008. 102(6): p. 669-76. 158. Johnstone, M.A., Pressure-dependent changes in nuclei and the process origins of

the endothelial cells lining Schlemm's canal. Invest Ophthalmol Vis Sci, 1979. 18(1): p. 44-51.

159. Xie, B., et al., Blockade of sphingosine-1-phosphate reduces macrophage influx

and retinal and choroidal neovascularization. J Cell Physiol, 2009. 218(1): p. 192-8.

160. Van Buskirk, E.M. and J. Brett, The canine eye: in vitro dissolution of the

barriers to aqueous outflow. Invest Ophthalmol Vis Sci, 1978. 17(3): p. 258-71. 161. Johnson, M., et al., The pressure and volume dependence of the rate of wash-out

in the bovine eye. Curr Eye Res, 1991. 10(4): p. 373-5. 162. Erickson-Lamy, K., et al., Absence of time-dependent facility increase

("washout") in the perfused enucleated human eye. Invest Ophthalmol Vis Sci, 1990. 31(11): p. 2384-8.

163. Vinken, M., et al., Involvement of cell junctions in hepatocyte culture

functionality. Crit Rev Toxicol, 2006. 36(4): p. 299-318.