Fisiopatologia de IRA Isquemica

download Fisiopatologia de IRA Isquemica

of 12

Transcript of Fisiopatologia de IRA Isquemica

  • 8/3/2019 Fisiopatologia de IRA Isquemica

    1/12NATURE REVIEWS|NEPHROLOGY VOLUME 7 | APRIL 2011 | 189

    Division of Nephrology,Department ofMedicine, IndianaUniversity School ofMedicine, 950 WestWalnut Street, R2202,Indianapolis, IN 46202,USA (A. A. Sharfuddin,B. A. Molitoris).

    Correspondence to:B. A. [email protected]

    Pathophysiology of ischemic acute kidney

    injuryAsif A. Sharfuddin and Bruce A. Molitoris

    Abstract | Acute kidney injury (AKI) as a consequence of ischemia is a common clinical event leading to

    unacceptably high morbidity and mortality, development of chronic kidney disease (CKD), and transition

    from pre-existing CKD to end-stage renal disease. Data indicate a close interaction between the many cell

    types involved in the pathophysiology of ischemic AKI, which has critical implications for the treatment of

    this condition. Inflammation seems to be the common factor that links the various cell types involved in this

    process. In this Review, we describe the interactions between these cells and their response to injury following

    ischemia. We relate these events to patients who are at high risk of AKI, and highlight the characteristics

    that might predispose these patients to injury. We also discuss how therapy targeting specific cell types can

    minimize the initial and subsequent injury following ischemia, thereby limiting the extent of acute changes and,hopefully, long-term structural and functional alterations to the kidney.

    Sharfuddin, A. A. & Molitoris, B. A. Nat. Rev. Nephrol.7, 189200 (2011); published online 1 March 2011; doi:10.1038/nrneph.2011.16

    Introduction

    The kidney is comprised of heterogeneous ce popua tions that function together to perform a number oftighty controed and compex processes. Acute kidneyinjury (AKI) is a common cinica event that disruptsthis homeostasis, eading to unacceptaby high morbid ity and mortaity. One cause of AKI is ischemia, whichcan occur for a number of reasons, for exampe, withthe use of vasoconstrictive drugs or radiocontrast agents;

    hypotension inked to sepsis or bood oss after surgeryand trauma is aso a known cause of ischemia. The bodyis abe to adapt to a reduction in bood fow to a certaineve, but when deivery of oxygen and metaboic sub strates becomes inadequate, ceuar injury eads to organdysfunction. In this Review, we describe the interactionsbetween the various ce types and processes invoved inthe pathophysioogy of ischemic AKI. We aso outine anapproach that wi faciitate patient care and deveopmentof therapies for ischemic AKI in the future.

    Patients at high risk of ischemic AKI

    A cardiovascuar event can ead to ischemic AKI in

    any patient, but there are certain individuas who are atinherenty high risk of deveoping AKI foowing mid tomoderate reductions in kidney perfusion. Thakar et al.1were the first to emphasize the importance of under standing and quantifying this increased risk. Subsequentstudies have identified and vaidated cinica variabes,such as age, existing chronic kidney disease (CKD), and

    proteinuria, that contribute to this increased risk,25 andcinica staging systems, incuding the RIFLE system,have been deveoped to cassify patients according totheir risk or injury.6,7

    These parameters shoud be used together with tradi tiona biomarkers when evauating risk in a patientundergoing eective procedures or receiving therapiesthat potentiay reduce rena bood fow.8 Rather than

    a singe characteristic, patients often have mutipeassociated risk factors that resut in a cumuative risk ofdeveoping ischemic AKI (Box 1).1 Identifying patientsat risk is an essentia component of the medica workup,invoving both history taking and a physica examina tion, as this wi maximize care and reduce risk. Forexampe, improving the hemodynamic status of a patientis important to minimize AKI from nephrotoxic drugssuch as cispatin and aminogycosides, or with the useof radiocontrast agents that reduce kidney perfusion.9Additiona factors, such as high eves of oxidativestress or infammation, aso increase a patients risk ofdeveoping AKI.10

    Prerena azotemia is the most common cause of AKIand accounts for 4055% of a cases.1113 This condi tion resuts from kidney hypoperfusion owing to areduced effective arteria voume, that is, the voumeof bood effectivey perf using the organs. Prerenaazotemia is divided into voume responsive and voumenonresponsive forms. In patients with the voume non responsive form, additiona intravascuar voume doesnot restore kidney perfusion and function. For exampe,patients with conditions such as congestive heart faiureand sepsis might not respond to intravenous fuid therapy,as decreased cardiac output and tota vascuar resistanceprevent kidney perfusion.1416 Compensation for prerena

    Competing interests

    B. A. Molitoris declares associations with the followingcompanies: Eli Lilly and Quark Pharmaceuticals. See the articleonline for full details of the relationships. A. A. Sharfuddindeclares no competing interests.

    FOCUS ON AKI IN CRIT ICAL CARE

    2011 Macmillan Publishers Limited. All rights reserved

    mailto:[email protected]://www.nature.com/doifinder/10.1038/nrneph.2011.16http://www.nature.com/doifinder/10.1038/nrneph.2011.16mailto:[email protected]
  • 8/3/2019 Fisiopatologia de IRA Isquemica

    2/12190 | APRIL 2011 | VOLUME 7 www.nature.com/nrneph

    azotemia incudes activation of baroreceptors, which ini tiates a cascade of neura and humora responses, activa tion of the sympathetic nervous system, and an increasein the production of catechoamines, especiay norepi nephrine.17 Increased reease of vasopressin is mediatedby hypovoemia and a rise in extraceuar osmoa ity, resuting in vasoconstriction, water retention, and

    Key points

    During ischemic acute kidney injury (AKI), ATP depletion results in cytoskeletal

    changes in epithelial and endothelial cells, causing disruption of function, and a

    decrease in glomerular filtration rate

    Apoptosis and necrosis are major mechanisms of cell death that have

    important roles in ischemia, with the contribution of each pathway depending

    on the extent of the injury

    Under physiological conditions, endothelial cells regulate permeabili ty, vascular

    tone, coagulation, and inflammation; endothelial cells that are dysfunctionalsubstantially contribute to the extension phase of AKI

    Inflammation and its mediators orchestrate the extension phase of ischemicAKI, and limit injury to tubular epithelial cells and vascular endothelial cells,

    thereby promoting repair

    Complex interactions between epithelial cells, endothelial cells, inflammatory

    mediators, and cytokines can result in persistent injury during acute tubular

    necrosis

    Stem cells, mesenchymal cells, and endothelial progenitor cells contribute to

    the repair and regeneration of tubular cells and endothelial cells following injury,

    and could provide attractive targets for therapeutic intervention

    back diffusion of urea into the papiary inter stitium.Activation of the reni nangiotensin adosteronesystem eads to increased production of the potent vaso constrictor angiotensin II, which preferentiay increasesefferent arterioar resistance.18 Gomeruar fitration rate(GFR) is preserved by the resutant increase in gomeru ar hydrostatic pressure. Angiotensin II activity is asoincreased during severe voume depetion, which eadsto afferent arterioar constriction, reducing rena pasmafow, GFR, and the fitration fraction.19

    Concomitanty, compensatory mechanisms of kidneyautoreguation preserve gomeruar perfusion.20 Underphysioogica conditions, autoreguation of rena boodfow works above a mean systemic arteria bood pres sure of 7580 mmHg. Beow this pressure, the gomeru ar utrafitration pressure and GFR decine abrupty.21Rena production of prostagandins, kaikrein, kinins,and nitric oxide (NO) are increased and contribute tovasodiation.22,23 Nonsteroida anti infammatory drugsinhibit the production of prostagandins and reducekidney perfusion. Angiotensin converting enzyme

    inhibitors bock the synthesis of angiotensin II anddisturb the deicate baance between afferent and effer ent arterioar tone in patients who have severe reductionsin effective arteria voume, such as those with congestiveheart faiure or biatera rena artery stenosis, thus wors ening prerena azotemia. Conversey, very high eves ofangiotensin II, as seen in circuatory shock, cause con striction of both afferent and efferent arterioes, whichnegates its protective effect.24

    Taken together, these data provide strong evidence thatcertain identifiabe patients are at high risk of deveopingAKI. Increased attention to maximizing conditions, suchas hemodynamic status, drug dosing based on GFR, and

    voume status is essentia to prevent or ameiorate AKIin these patients.

    Cellular changes during ischemic AKI

    Acute epithelial cell injury

    Foowing a reduction in effective kidney perfusion,epitheia ces are unabe to maintain adequate intra ceuar ATP for essentia processes. This depetion ofATP eads to ce injury and, if severe enough, ce deathby necrosis or apoptosis. A segments of the nephroncan be affected during an ischemic insut, but the mostcommony injured epitheia ce is the proxima tubuarce. These ces are particuary susceptibe for a number

    of reasons. First, this ce type has a high metaboic raterequired for mediating ion transport and a imited cap acity to undergo anaerobic gycoysis. Second, owingto the unique bood fow in the outer stripe of the S3segment of the nephron, there is marked microvascuarhypoperfusion and congestion in this region after injury,which persists and mediates continued ischemia evenwhen cortica bood fow might have returned to near norma eves. Endotheia ce injury and dysfunctionare primariy responsibe for this phenomenon, knownas the extension phase of AKI.25 Understanding that isch emic injury can be ocaized to specific microvascuardomains rather than throughout the kidney is important,

    Box 1 | Causes of reduced effective ar terial volume and kidney hypoperfusion

    Intravascular volume depletion

    Hemorrhage (e.g. following trauma, surgery, postpartum)

    Gastrointestinal losses (e.g. from diarrhea, vomiting, nasogastric loss)

    Renal losses (e.g. diuretics, osmotic diuresis, diabetes insipidus)

    Skin and mucous membrane losses (e.g. burns, hyperthermia)

    Nephrotic syndrome

    Cirrhosis

    Capillary leak

    Reduced cardiac output

    Cardiogenic shock

    Pericardial disease (e.g. restrictive, constrictive, tamponade)

    Congestive heart failure

    Valvular heart disease

    Pulmonary disease (e.g. pulmonary hypertension, pulmonary embolism)

    Sepsis

    Systemic vasodilation

    Cirrhosis

    Anaphylaxis

    Sepsis

    Renal vasoconstriction

    Early sepsis

    Hepatorenal syndrome

    Acute hypercalcemia

    Molecules and drugs (e.g. norepinephrine, vasopressin, nonsteroidal anti-

    inflammatory drugs, angiotensin-converting enzyme, calcineurin inhibitors)

    Radiocontrast agents

    REVIEWS

    2011 Macmillan Publishers Limited. All rights reserved

  • 8/3/2019 Fisiopatologia de IRA Isquemica

    3/12NATURE REVIEWS|NEPHROLOGY VOLUME 7 | APRIL 2011 | 191

    as quantifying tota rena bood fow as a measure ofeffective bood fow coud be miseading.

    The other major epitheia ces of the nephron invovedin the pathophysioogy of ischemic AKI are those ofthe meduary thick ascending imb ocated distay.Apoptotic changes have been detected in human AKI,as shown in dista nephron segments during nephro toxic acute tubuar necrosis. Apoptosis of dista tubuarces aso occurs in donor biopsies before engraftment,which was predictive in one study of deayed graft func tion.26 In an ex vivo mode of hypoxic AKI, administra tion of FG 4497 (a specific proy hydroxyase inhibitorthat activates hypoxia inducibe factor [FibroGen, SanFrancisco, CA, USA]) in the isoated perfused kidneyed to decreased seective outer meduary dista tubuarinjury.27 Proxima tubuar ce injury and dysfunctionduring ischemia or sepsis eads to afferent arterioarvasoconstriction mediated by tubuogomeruar feed back, umina obstruction, and backeak of fitrateacross injured proxima tubuar ces, resuting inineffective gomeruar fitration and a profound drop

    in GFR (Figure 1).28,29

    Morphological changes

    A hamark of ischemic injury is oss of the apica brushborder of proxima tubuar ces. Disruption of microviiand their detachment from the apica ce surface eadsto formation of membrane bound bebs eary foow ing ischemia that are reeased into the tubuar umen.Detachment and oss of tubuar ces exposes areasof denuded basement membrane, resuting in foca areas ofproxima tubuar diatation, as we as formation of distatubue casts.30 The soughed tubuar ces, brush bordervesice remnants, and ceuar debris in combination with

    uromoduin form these granuar casts, which have thepotentia to obstruct the tubue umen, eading to noGFR in that functiona unit.31 Necrotic ce death is rareand restricted to the highy susceptibe outer meduaryregions, whereas features of apoptosis are commony seenin both proxima and dista tubuar ces (see beow).32

    Injured gomeruar epitheia ces foowing isch emic or septic injury are not usuay seen on histoogicastains, athough studies have shown reversibe podocyte specific moecuar and ceuar changes. Wagner et al.33demonstrated in a rat mode that rena ischemia inducedpodocyte effacement with oss of sit diaphragm integ rity and proteinuria owing to rapid oss of interactions

    between the tight junction proteins Neph1 and ZO 1.Ce cuture modes using human podocytes showed thatATP depetion resuted in rapid oss of Neph1 and ZO 1binding, and redistribution of Neph1 and ZO 1 from thece membrane to cytopasm; ATP recovery increasedphosphoryation of Neph1 and restored Neph1 and ZO 1binding and their ocaization at the ce membrane.33

    Cytoskeletal and structural changes

    The actin cytoskeeton has an integra roe in maintain ing ce structure and function, poarity, endocytosis,signa transduction, motiity, movement of organees,exocytosis, ce division, migration, barrier function of

    the junctiona compexes, and cematrix adhesion.34Maintaining the integrity of the cytoskeeton is especiay

    important for proxima tubuar ces in which ampifica tion of the apica membrane by microvii is essentia fornorma ce function. Depetion of ceuar ATP eadsto rapid disruption of apica F actin by depoymeriza tion mediated in part by cofiin, and redistribution ofthe cytoskeeta F actin core. This disruption causesinstabiity of the surface membrane and formation ofmembrane bound extraceuar vesices or bebs that areeither exfoiated into the tubuar umen or internaizedto potentiay be recyced.3539 Other proteins invoved inthe depoymerization process are tropomyosin and ezrin.During ischemia, ezrin becomes dephosphoryated andthe attachment between the microviar F actin core

    and pasma membrane is ost.40 Simiary, tropomyosinsbind to and stabiize the F actin microfiament core in thetermina web by preventing access to cofiin. Foowingischemia, there is dissociation of tropomyosins from themicrofiament core, which enabes access of the micro fiaments in the termina web to the binding, severing,and depoymerizing actions of cofiin.41

    Another important consequence of disruption ofthe actin cytoskeeton is the oss of tight junctions andadherens junctions. These junctiona compexes activeyparticipate in numerous functions, such as paraceuartransport, ce poarity, and ce morphoogy. Eary isch emic injury resuts in opening of tight junctions, eading

    Endothelial injury

    ActivationDysfunctionDetachmentApoptosisNecrosis

    Inammation

    WBC recruitmentNeutrophils

    MacrophagesLymphocytesDendritic cell

    activation

    Vasoconstriction

    Cytokine releasePermeability

    Leukocyte adhesionmolecules

    Rouleaux formationReduced ow

    Leukocyte activation

    Cytokine releaseMargination

    Tissue migrationReduced ow

    Cellular sheddingCellular debrisLoss of polarity

    Loss of tight junctionsCytokine release

    Epithelial cell injury

    Sub-lethal injury Cytoskeleton

    disruption

    Lethal injury Necrosis Apoptosis

    TubularobstructionBackleak

    TGF-

    Defective functionReduced GFR

    High FENaConcentrating defect

    Figure 1 | Pathogenesis of ischemic AKI. The major pathways of GFR impairmentin ischemic acute tubular injury are caused by ATP depletion in vascular andtubular cells. Numerous interactions exist between endothelial cells, WBCs,and epithelial cells in the pathophysiology of ischemic AKI. These interactions arebidirectional between the cells involved, and result in specific functional and

    structural alterations. Inflammatory mediators released from proximal tubular cellsinfluence endothelial cell processes (e.g. increase vasoconstriction andexpression of cell adhesion molecules) that in turn influence the interactionsbetween WBCs and endothelial cells, leading to reduced microvascular flow andcontinued hypoxia within the local environment. Additional functional changesoccur, such as a marked reduction in production of erythropoietin and25-hydroxylation of vitamin D. Electrolyte accumulation can rapidly lead torequirement of renal replacement therapy. Metabolic acidosis as a consequence ofAKI must also be carefully monitored. Abbreviations: AKI, acute kidney injury;FENa, fractional excretion of sodium; GFR, glomerular filtration rate; TGF-,transforming growth factor ; WBC, white blood cell.

    FOCUS ON AKI IN CRIT ICAL CARE

    2011 Macmillan Publishers Limited. All rights reserved

  • 8/3/2019 Fisiopatologia de IRA Isquemica

    4/12192 | APRIL 2011 | VOLUME 7 www.nature.com/nrneph

    to increased paraceuar permeabiity and backeak ofthe gomeruar fitrate into the interstitium.42 Duringischemia, epitheia ces aso ose their attachment tothe underying extraceuar matrix owing to disruptionof integrins. Depetion of ATP resuts in reocaization of integrins from the basa membrane to the apica mem brane, with subsequent detachment of viabe ces fromthe tubuar basement membrane.43 The exfoiated cesthen bind to each other and form ceuar casts withinthe tubuar umen.

    Aterations to the actin cytoskeeton during ischemiaresut in changes in ce poarity and function (Figure 2).Basoatera Na+/K+ATPase pumps redistribute to theapica membrane as eary as within 10 min foowingdisruption of the spectrinactin cytoskeeton, which isresponsibe for attaching the pumps to the membrane.44Redistribution of the pumps resuts in bidirectionatransport of sodium and water across the apica, as weas the basoatera, epitheia ce membrane, with ceuarsodium being transported back into the tubuar umen.This process is one the major mechanisms of the highfractiona excretion of sodium seen in patients with acutetubuar necrosis, and the inefficient use of ceuar ATP,

    as ischemic conditions uncoupe ATP use and effec tive transceuar sodium transport.45 A high sodiumconcentration in the fitrate reaching the dista tubueeads to a reduction in GFR by activation of tubuargomeruar feedback, with stimuation of the macuadensa mediating afferent arterioar vasoconstriction.

    Apoptosis and necrosis

    The fate of the epitheia ce foowing an ischemicevent utimatey depends on the extent of injury. Cesundergoing sub etha or ess severe injury have the cap abiity of functiona and structura recovery if the insutis interrupted. Ces that suffer a more severe or ethainjury undergo apoptosis or necrosis, eading to cedeath. Apoptosis is an energy dependent, programmedce death that resuts in condensation of nucear andcytopasmic materia to form apoptotic bodies. Thesemembrane bound apoptotic bodies are rapidy phago cytosed by macrophages and neighboring epitheia ces.During necrosis, there is ceuar and organee sweing,oss of pasma membrane integrity, and rapid reease of

    cytopasmic and nucear materia into the umen or inter stitium.46 Secondary necrosis occurs when ces under going apoptosis do not have adequate ceuar eves ofATP to support the staged demise of the ce.

    Apoptotic mechanisms are compex with factorsaffecting a number of pathways. The caspase famiy ofproteases is an important initiator and effector of apop tosis.47,48 Both intrinsic (mitochondria) and extrinsic(death receptor) apoptotic pathways are activated inAKI. Specificay, activation of procaspase 9 primariydepends on intrinsic mitochondria pathways reguatedby the Bc 2 famiy of proteins, whereas pro caspase 8activation resuts from extrinsic signaing via ce surface

    death receptors, such as FAS and FADD.47,48 Considerabecrosstak aso exists between the intrinsic and extrinsicpathways. Caspase 3, caspase 6, and caspase 7 are effec tor caspases that are abundant and catayticay robust,ceaving many ceuar proteins, which resuts in thecassic apoptotic phenotype. Inhibition of caspase activ ity has been shown to be protective against injuryin vitroand in AKI in vivo.49,50

    Severa apoptotic pathways, incuding the intrinsic(Bc 2 famiy, cytochrome c, caspase 9), extrinsic (FAS,FADD, caspase 8) and reguatory (p53 and nucearfactor B) pathways, seem to be activated during isch emic rena tubuar ce injury. The baance between

    ce surviva and apoptotic ce death aso depends onthe reative concentrations of the proapoptotic (BAX,BAD, and BID) and antiapoptotic (Bc 2 and B c 2 ike protein 1) members of the Bc 2 famiy of proteins.Overexpression of proapoptotic or reative deficiencyof antiapoptotic proteins can ead to formation of mito chondria pores. Other proteins that have an importantroe in apoptotic pathways incude nucear factor Band p53.51,52 The centra proapoptotic transcriptionfactor p53 can be activated by hypoxia, via hypoxia inducibe factor, as we as other noxious stimui suchas certain drugs. Kinases are responsibe for mediat ing ceuar responses invoved in apoptosis, surviva,

    Brush border

    Blebbing of microvilli

    Injury Recovery

    Tight junctions

    Basement membrane

    Loss of actin

    cytoskeletal structure

    Cast formation

    Loss of cellcell contacts

    Redistribution of

    Na+/K+-ATPases

    and integrins

    to apical location

    Epithelial

    cell swelling

    Backleak

    Actin cytoskeleton

    Cell adhesionmolecules

    Integrins

    Na+/K+-ATPase

    Figure 2 | Effects of sub-lethal injury to tubular cells and their recovery. Damage toepithelial cells occurs early during ischemia and involves alterations to thecytoskeleton and in surface membrane polarity. ATP depletion induces rapiddisorganization of the actin cytoskeleton structure, which disrupts tight junctionsand in turn leads to backleak of tubular filtrate. Loss of cellcell contacts and celladhesion molecules results in flattened nonpolarized epithelial cells, denudedbasement membranes, and expression of mesenchymal markers. Na+/K+-ATPasepumps normally located at the basolateral membrane and tethered by the actinspectrin cytoskeleton, redistribute to the apical membrane of the proximal tubularcell and are internalized into the cytosol during ischemic injury. Morphologically,proximal tubular cells lose their brush borders, undergo swelling, and blebbing ofmicrovilli during injury, leading to cast formation. Severely injured proximal tubularcells undergo mesenchymal differentiation and subsequent re-epithelization.Recovery of proximal tubular cells begins with integrin reattachment, reassembly ofthe actin cytoskeleton, repolarization of the surface membranes, and redistribution

    of the sodium pumps back to their basolateral location.

    REVIEWS

    2011 Macmillan Publishers Limited. All rights reserved

  • 8/3/2019 Fisiopatologia de IRA Isquemica

    5/12NATURE REVIEWS|NEPHROLOGY VOLUME 7 | APRIL 2011 | 193

    and repair through their interaction with signas fromgrowth factors, incuding hepatocyte growth factor,insuin ike growth factor I, epiderma growth factor, andvascuar endotheia growth factor (VEGF).53,54 Thesemechanisms, which can be activated independenty vianonischemic pathways in other types of injury, inhibitproapoptotic proteins and activate the antiapoptotictranscription of cycic AMP response eement bindingfactors. Knock down of p53 in proxima tubuar cesusing short interfering RNA ed to a dose dependentattenuation of apoptotic signaing and kidney injury incamp ischemia and transpant modes, indicating poten tia therapeutic benefit for ischemic and nephrotoxickidney injury.55,56

    Necrosis of epitheia ces resuts from increasedintraceuar cacium and the activation of membranephosphoipases and capain.57,58 Necrotic ces do not,therefore, exhibit the nucear fragmentation or chro matin condensation seen in apoptosis, and neither dothey form apoptotic bodies. Functionay, severe ATPdepetion resuts first in mitochondria injury with sub

    sequent arrest of oxidative phosphoryation, causingfurther depetion of energy stores and robust forma tion of reactive oxygen species. Reactive oxygen species,such as the hydroxy radica peroxynitrite and hyper chorous acid, are generated by cataytic conversion inepitheia ces during ischemic injury. These moecuesdamage ces in a variety of ways, incuding peroxidationof ipids in the pasma membrane and intraceuar mem branes, and destabiization of cytoskeeta proteins andintegrins required to maintain cece adhesion as weas interactions between ces and the extraceuar matrix.Reactive oxygen species can aso have vasoconstrictiveeffects by scavenging NO.59

    Autophagy, which is the process invoved in degrada tion of a ces own components through the ysosomamachinery, is now increasingy recognized as perhaps themost frequent ce death pathway for injured epitheium.Li et al.60 demonstrated the important roe of autophagyin rena epitheia ces in obstructive uropathy modes,whie Koesters et al.61 showed that expression of trans forming growth factor eads to excessive autophagy ininjured tubues.

    Endothelial dysfunction

    Endotheia ces contribute to vascuar tone, reguationof bood fow to oca tissue beds, moduation of coagua

    tion and infammation, and vascuar permeabiity. Bothischemia and sepsis have profound effects on the renaendotheium, resuting in microvascuar dysreguationand continued ischemia and further injury, especiay inthe outer stripe of the kidney. Histopathoogica examina tion shows vascuar congestion, formation of edema,diminished microvascuar bood fow, and marginationand adhesion of infammatory ces to the endotheium,eading to the extension phase of AKI.34 Athough amarked decrease in tota kidney perfusion resuts ingoba ischemia, decreased regiona perfusion can extendischemic injury ocay without affecting goba perfu sion. The compexity of the vascuar beds within the

    kidney makes interpretation of tota kidney bood fowchaenging foowing ischemic or septic injury.

    Vascular tone

    Conger et al.62,63 were among the first to demonstratethat postischemic rat kidneys dispayed vasoconstrictionin response to decreased rena perfusion pressure and,therefore, coud not autoreguate bood fow, even whentota rena bood fow had returned to baseine vaues upto 1 week after injury. This response can be bocked byCa2+ antagonists, and oss of endotheia NO synthase(eNOS) function is due to a reduced vasodiator responseto acetychoine and bradykinin.6466 Seective inhibition,depetion, or deetion of inducibe NOS (iNOS) showsrenoprotective effects during ischemia.64,65 Overa, thereis an imbaance of eNOS and iNOS in ischemic AKI, andit has been proposed that owing to a reative decreasein eNOS, secondary to endotheia dysfunction anddamage, there is a oss of antithrombogenic propertiesof the endotheium eading to increased susceptibiity tomicrovascuar thrombosis.66

    Administration of l arginine, the NO donor mo sidomine, or the eNOS co factor sapropterin can pre serve meduary perfusion and attenuate AKI induced byischemiareperfusion injury; conversey the administra tion ofNnitro l arginine methy ester, an NO bocker,has been reported to aggravate the course of AKI.67 Severapharmacoogica studies have assessed the contribution ofeNOS impairment to the overa course of reduced renafunction foowing ischemiareperfusion injury.68,69

    Permeability

    The endotheia barrier separates the umen of the boodvesse from the surrounding tissue, and contros the

    exchange of ces and fuid between these two compart ments. The endotheium is defined by transceuar andparaceuar pathways, the atter being a major contribu tor to endotheia dysfunction induced by infammation.Sutton et al.70 studied the roe of endotheia ces in AKIby utiizing fuorescent dextrans and two photon intra vita imaging in a series of experiments. The increasedmicrovascuar permeabiity observed in AKI is ikey tobe caused by a combination of factors, such as disrup tion of the endotheia monoayer and actin cytoskeeton,breakdown of perivascuar matrix, aterations in contactsbetween endotheia ces, upreguated eukocyteendo theia interactions, and severe aterations in the integrity

    of the adherens junctions of the rena microvascuature.In vivo two photon imaging demonstrated a oss ofcapiary barrier function within 24 h of reperfusion,with maxima effects seen at 24 h after injury.70

    Breakdown of the barrier function provided by theendotheium might aso be due to activation of matrixmetaoproteinase 2 or matrix metao proteinase 9,which temporay correates with an increase in micro vascuar permeabiity.25,71 Minocycine, a broad spectruminhibitor of matrix metaoproteinases, and the geatinaseinhibitor ABT 518 (Abbott, Abbott Park, IL, USA), bothameiorated the increase in microvascuar permeabiityin a rat mode of ischemic rena injury.71

    FOCUS ON AKI IN CRIT ICAL CARE

    2011 Macmillan Publishers Limited. All rights reserved

  • 8/3/2019 Fisiopatologia de IRA Isquemica

    6/12194 | APRIL 2011 | VOLUME 7 www.nature.com/nrneph

    Coagulation

    Endotheia ces have a centra roe in coaguation

    through their interaction with protein C and thrombo moduin. Protein C is activated by thrombin mediatedceavage and the rate of this reaction is augmented1,000 fod when thrombin binds to the endotheiace surface receptor thrombomoduin.72 The activa tion rate of protein C is further increased by approxi matey 10 fod when endo theia protein C receptor(EPCR) binds protein C and presents it to the thrombinthrombomoduin compex. Activated protein C acquiresantithrombotic and profibrinoytic properties, and partici pates in numerous anti infammatory and cytoprotectivepathways to restore norma homeostasis.73 Activatedprotein C is aso an agonist of proteinase activated

    receptor 1.74During an infammatory response, many of the

    natura anticoaguants, incuding protein C, aredegraded, or their production is decreased togetherwith downreguation of EPCR and thrombomoduinexpression, which decreases the anticoaguant and anti infammatory effects of the protein C pathway. Damagedendotheia ces undergo apoptosis, which ampifiesthe coaguation cascade further by providing a pro coaguant surface.75 Continued activation of infamma tion and the coaguation pathway eads to increasedmicrovascuar coaguation and further endotheiace dysfunction. Utimatey, microvascuar function is

    compromised, resuting in disseminated intravascuarcoaguation and thrombosis, decreased oca tissue per

    fusion, and organ dysfunction or faiure. Treatment withthrombomoduin both before and after injury attenuatesdamage, with minimization of vascuar permeabiitydefects, and improved rena bood fow.76

    Activation of eukocytes and their reease of c yto kines requires signas from chemokines circuatingin the boodstream or through direct contact with theendotheium (Figure 3). Roing eukocytes can be acti vated by chemoattractants, such as compement C5a andpateet activating factor. Once activated, integrins on theeukocytes bind to endotheia igands to promote firmadhesion, with integrin 2 being the most important.77These interactions with the endotheium are medi

    ated through endotheia adhesion moecues that areupreguated during ischemic conditions.

    Singbart et al.78 found that P seectin on pateets,but not on endotheia ces, was the main determi nant in neutrophi mediated ischemic AKI. Bockadeof the common igand for E seectin, P seectin, andL seectin provided protection from both ischemicinjury and mortaity, which seemed to depend onthe presence of a key fucosy sugar on the s eectinigand.79,80 In a ceca igation and puncture (CLP) modeof septic peritonitis, mice engineered to be deficientfor E seectin, P seectin, or both, were competeyprotected against injury.81,82

    a

    Loss of endothelial

    cellcell contacts

    Coagulation

    Impaired ow

    Rouleauxformation

    Endothelial cellswelling

    Expression of

    adhesion molecules

    Transendothelial migration

    Interstitium

    Proximal tubuleCapillary

    Cytokinerelease

    Cytokines

    Chemokines

    ROS

    ECMbreakdown

    Permeability

    Cytokines

    DCLeukocyteendothelial

    cell adhesion and

    interaction

    Lumen

    b

    Figure 3 | Events in endothelial cell activation, injury, and reduced microvascular flow. a | Ischemia causes upregulation andexpression of genes encoding various cell surface proteins, such as E-selectin, P-selectin, vascular cell adhesion protein 1,

    and intercellular adhesion molecule 1, and downregulation of thrombomodulin. Activated leukocytes bind to endothelialcells through these adhesion molecules. Endothelial injury increases the production of endothelin-1 and decreasesendothelium-derived nitric oxide synthase, which induces vasoconstriction and platelet aggregation, promoting ahypercoagulable environment. The combination of leukocyte adhesion and activation, platelet aggregation, and endothelialinjury serves as the basis for vascular congestion of the cortical and medullary microvasculature. Permeability defectsbetween endothelial cells occur as a result of alterations in tight junctions and adherens junctions. The close proximity andcrosstalk between the epithelial proximal tubular cells and microvascular endothelial cells, as well as release of cytokinesand chemokines, further increase inflammation. Dendritic cells also have a role in this inflammatory cascade, and amplifyinflammatory signals between endothelial cells and epithelial cells. b | Hematoxylin and eosin stain of a human kidneybiopsy from a patient with AKI following ischemic injur y. Abbreviations: AKI, acute kidney injury; DC, dendritic cell; ECM,extracellular matrix; ROS, reactive oxygen species.

    REVIEWS

    2011 Macmillan Publishers Limited. All rights reserved

  • 8/3/2019 Fisiopatologia de IRA Isquemica

    7/12NATURE REVIEWS|NEPHROLOGY VOLUME 7 | APRIL 2011 | 195

    Long-term effects of endothelial injury

    Injury to endotheia ces coud contribute to chronicdisease. Basie et al.83 documented a considerabedecrease in the density of bood vesses foowing acuteischemic injury, which ed to the phenomenon of vascu ar dropout. This phenomenon was verified by Hrbetet al.84 who found that vascuar density was reduced byamost 45% at 4 weeks after an ischemic insut. Thisobservation indicates that, unike rena epitheia tubuarces, the rena vascuar system acks comparabe regener ative potentia. Whether apoptosis and necrosis contrib ute to vascuar dropout is not yet cear. Ischemia has beenshown to inhibit VEGF, whie inducing the VEGF inhibi tor ADAMTS1.85 The ack of vascuar repair was postu ated to be due to the reduction in VEGF expression, asadministration of VEGF to postischemic rats preservedmicrovascuar density.86 Vascuar dropout might mediateincreases in the expression of hypoxia inducibe factorand fibrosis, and ater proper hemodynamics, eading tohypertension. Basie and co workers have shown thatthe poor regenerative potentia of endotheia ces and

    transformation into fibrobasts is in arge part owingto the ack of VEGF expression,87 which coud acceeratethe progression of CKD foowing initia recovery fromischemiareperfusion injury induced AKI. 83,88 Vascuardropout coud predispose individuas to recurrentischemic events and AKI.9

    Leukocytes and inflammation

    Infammation and recruitment of eukocytes during epi theia injury are now recognized as major mediators ofa phases of endotheia and tubuar ce injury (Figure 3).Eary infammation is characterized by margination ofeukocytes to the activated vascuar endotheium via

    interactions between seectins and igands that enabefirm adhesion, foowed by transmigration into the inter stitium. A number of potent mediators are generatedby the injured epitheia proxima tubuar ce, incud ing proinfammatory cytokines, such as tumor necrosisfactor (TNF), intereukin (IL) 6, IL 1, IL 8, C C motifchemokine 2, transforming growth factor , and C Cmotif chemokine 5.89 To ike receptor (TLR) 2 is animportant mediator of endotheia ischemic injury, whieTLR4 has been shown in anima modes of both ischemicand septic injury to aso have a roe in injury,90 especiayin proxima tubuar ces.91

    Neutrophis are the first ces to accumuate at the site

    of ischemic injury.91 Bockade of neutrophi functionor neutrophi depetion provides ony partia protec tion against injury, indicating that other eukocytes asomediate injury. These infammatory mediators incudemacrophages, B ces, and T ces.92 Seective deetionof these ces in knockout mouse modes, and throughantibody mediated bockade, shows that these cesmediate tubuar injury at various phases of the process,and there are synergistic interactions between differ ent ce types.93 Expression of the compement compo nent C5a is markedy upreguated on proxima tubuarepitheia ces as we as interstitia macrophages, andis a powerfu chemoattractant with procoaguant

    properties.94 Compement cascades are activated duringsepsis, and C5a has been found to be eevated in rodentmodes of sepsis.95 Bocking C5a or its receptor coudimprove surviva in sepsis.95,96

    Thurman et al.97,98 showed that C3a was requiredfor CXC chemokine production by epitheia ces, andCrry (a compement inhibitor ocaized to the basoateramembrane of epitheia ces) was decreased foowingischemic injury. Knock down of C3a and caspase 3 usingRNA interference protected rena function in a transpantmode.99 Numerous cytokines, whether reeased from theendotheium or epitheia ces, work together to augmentthe infammatory response foowing ischemic or septicinjury.100 Cutured mouse tubuar ces stimuated withipopoysaccharide ed to an upreguation of TLR2, TLR3,and TLR4 and secretion of C C chemokines, such as C Cmotif chemokine 2 and C C motif chemokine 5. Thesedata indicate that expression of tubuar TLRs might beinvoved in mediating interstitia eukocyte infitrationand tubuar injury during bacteria sepsis.101

    TLR2 and TLR4 are constitutivey expressed on

    rena epitheium, and their expression is increased fo owing rena ischemiareperfusion injury. E Achkaret al.102 showed in a CLP rat mode of sepsis that Tr4expression was increased markedy in a tubues (proxi ma and dista), gomerui, and the rena vascuature.Furthermore, this group demonstrated that sepsis edto a Tr4 dependent increase in the expression of theproinfammatory mediator Cox 2; this protein wasmosty restricted to cortica and meduary thick ascend ing oops of Hene, which characteristicay express andsecrete uromoduin.103 Uromoduin coud stabiize theouter medua during injury by decreasing infammation,possiby through an effect on TLR4.104 Genetic deetion

    of either TLR2 or TLR4 protects against rena ischemiareperfusion injury,91,105 thus indicating the prominentroe of TLRs in AKI.

    Macrophages produce proinfammatory cytokines thatcan stimuate the activity of other eukocytes. Dayet al.106showed that depetion of macrophages in the kidney andspeen using iposoma codronate before rena ischemiareperfusion injury prevented AKI, whereas adoptivetransfer of macrophages reconstituted AKI. This groupaso demonstrated that agonists of the sphingosine 1 phosphate receptor (S1PR) induced ymphopenia, whichhad a protective effect.106,107 However, studies have asoshown a ymphocyte independent roe of S1PR in main

    taining structura integrity after AKI, as S1PRs in theproxima tubue are necessary for stress induced cesurviva, and agonists of this receptor are renoprotectivevia direct effects on tubuar ces.108 Dendritic ces areaso thought to have a roe in AKI. Dong et al.109 demon strated that after AKI, rena dendritic ces producethe proinfammatory cytokines TNF, IL 6, C C motifchemokine 2, and C C motif chemokine 5, and thatdepetion of dendritic ces before ischemia substantiayreduced the eves of TNF produced by the kidney.

    T reguatory (TREG

    ) ces aso have a roe in ischemicAKI. Gandofo et al.110 showed in a murine mode of isch emic AKI that T

    REGce trafficking into the kidneys was

    FOCUS ON AKI IN CRIT ICAL CARE

    2011 Macmillan Publishers Limited. All rights reserved

  • 8/3/2019 Fisiopatologia de IRA Isquemica

    8/12196 | APRIL 2011 | VOLUME 7 www.nature.com/nrneph

    increased after 3 days and 10 days. Postischemic kidneyshad increased numbers of T ce receptor (TCR) +CD4+and TCR+CD8+ T ces, with increased production ofproinfammatory cytokines. The researchers noted thatdepetion of T

    REGces using anti CD25 anti body 1 day

    after ischemic injury increased rena tubuar damage,reduced tubuar proiferation, increased the productionof cytokines from infitrating T ces at 3 days, and TNFgeneration by TCR+CD4+ T ces at 10 days. In a separatestudy, infusion of CD4+CD25+ T

    REGces 1 day after initia

    injury reduced production of interferon by TCR+CD4+T ces at 3 days, improved repair, and reduced cytokinegeneration at 10 days.111 These studies demonstrate thatT

    REGces infitrate reperfused kidneys during the heaing

    process, promoting repair, ikey through moduation ofproinfammatory cytokine production of other T cesubsets. Partia depetion of T

    REGces with an anti

    CD25 monocona antibody potentiated kidney damageinduced by ischemiareperfusion injury, and resutedin more neutrophis, macrophages, and innate cytokinetranscription in the kidney.111 Furthermore, mice

    deficient in Foxp3+ TREG ces had a greater accumua tion of infammatory eukocytes after rena ischemiareperfusion injury than mice containing T

    REGces;

    co transfer of isoated TREGS

    ces and Scurfy ymph nodeces attenuated ischemiareperfusion injury inducedrena injury and eukocyte accumuation.112

    Natura kier ces have been reported to infitrate thepostischemic kidney by 4 h after reperfusion. Li et al.113demonstrated the essentia roe of natura kier cesand neutrophis in the innate immune response to renaischemiareperfusion injury by mediating neutrophiinfitration and production of interferon . Furthermore,considerabe protection from kidney ischemia

    reperfusion injury was evident in mice deficient in naturakier ces and mice administered an anti CD1d mono cona antibody, which bocked the interaction betweenantigen presenting ces and natura kier ces. 113

    The anticoaguant function of antigen presentingces is responsibe for suppressing ipopoysaccharide induced stimuation of the proinfammatory media tors angiotensin converting enzyme 1, IL 6, and IL 18,perhaps accounting for its abiity to moduate renahemodynamics and protecting against septic AKI.114Taken together, these findings show that suppressionof infammation is a key target towards preventing andimiting AKI.

    Investigators have shown that T ces aso have a majorroe in vascuar permeabiity during ischemic injury.Gene microarray anaysis showed that the productionof TNF and interferon was increased in CD3 andCD4 T ces from the bood and kidney after ischemia.Furthermore, it has aso been demonstrated that, in micedeficient in T ces expressing CD3, CD4, and CD8, thereis an attenuated increase in rena vascuar permeabiityafter ischemic injury. In this way, T ces directy con tribute to the increased vascuar permeabiity, potentiaythrough production of cytokines.115,116

    Another feature noted during infammation andendotheia ce injury is the phenomenon of erythrocyte

    trapping with Roueaux formation, causing obstructionand proonging the reduction in microvascuar boodfow and exacerbating tubuar injury.35

    Distant organ effects of AKI

    Ischemic AKI can have distant effects that potentiayater the function of other organs. Keyet al.117 demon strated the effects of rena ischemia on cardiac tissuesas shown by induction of IL 1, TNF, and interceuaradhesion moecue 1 mRNA expression as eary as 6 hafter ischemia. Kramer et al.118 showed that rena isch emic injury ed to an increase in pumonary vascuarpermeabiity defects, which were mediated throughmacrophages. Furthermore, this group showed in a ratmode of biatera rena ischemic injury or nephrectomythat expression of ung epitheia sodium channe, Na+/K+ATPase, and aquaporin 5 were downreguated, whichwas not the case in uniatera ischemic modes, indicat ing a roe for uremic toxins in moduating these effectsin the ung.119 Functiona changes in the brain have asobeen shown in the setting of AKI as noted in mice that

    had increased neurona pyknosis and microgiosis inthe brain.120 In addition, extravasation of Evans bue dyeinto the brain indicated that the boodbrain barrier wasdisrupted in mice with AKI.120

    Extrarena organs might conversey reguate isch emic AKI. Traumatic brain injury eicits a cytokine andinfammatory response that eads to rena infammationin transpants from brain dead, but not iving donors. 121The fact that AKI is associated with high morbid ity and mortaity indicates that mutiorgan crosstakis ikey to be a major contributor to dysfunction ofnonrena organs.

    Molecules that protect against injuryMuch of the discussion above has focused on proteins orevents that promote injury. However, there are protec tive mechanisms that provide a defense against numer ous stresses. The heat shock protein system is inducedduring stress conditions; for exampe, overexpressionof heat shock proteins 25, 90, and 72 before injury havebeen found to have protective effects.122124 These pro teins are beieved to hep restore norma ce function byassisting in the refoding of denatured proteins, as weas aiding the appropriate foding of newy synthesizedproteins. Heat shock proteins aso degrade irreparabeproteins and toxins to imit their accumuation.

    The enzyme heme oxygenase 1 has anti infammatory,vasodiatory, cytoprotective, antiapoptotic, and anti proiferative effects.125127 Mice deficient in this enzymewere shown to have marked exacerbation of gycero induced AKI, whereas overexpression of heme oxy genase 1 in cutured rena epitheia ces inducedupreguation of the ce cyce inhibitory protein p21,which conferred resistance to apoptosis.125127 Therefore,the actions of heme oxygenase 1 make it a potentiaytherapeutic enzyme in the prevention and reductionof AKI. More importanty, the upreguation or over expression of heme oxygenase 1 might aso be beneficiain the repair and regeneration of tubuar ces.

    REVIEWS

    2011 Macmillan Publishers Limited. All rights reserved

  • 8/3/2019 Fisiopatologia de IRA Isquemica

    9/12

  • 8/3/2019 Fisiopatologia de IRA Isquemica

    10/12198 | APRIL 2011 | VOLUME 7 www.nature.com/nrneph

    where they ameiorate AKI through both paracrineeffects as we as repair of the injured microvascuature.146Human HSCs administered systemicay 24 h after kidneyinjury were seectivey recruited to injured kidneys ofimmunodeficient mice and ocaized prominenty inand around the vascuature.147 This recruitment wasassociated with repair of the kidney microvascuatureand tubuar epitheia ces, improved functiona recov ery, and increased surviva. HSCs recruited to the kidneyexpressed markers consistent with circuating endotheiaprogenitor ces and synthesized high eves of proangio genic cytokines, which promoted proiferation of bothendotheia and epitheia ces. Athough purified HSCsacquired endotheia progenitor markers once recruitedto the kidney, engraftment of human endotheia ces inthe mouse capiary was was rare, indicating that renarepair by human stem ces is mediated by paracrinemechanisms rather than repacement of the vascua ture.147 Targeting mechanisms of injury in this way tobock dysfunctiona intraceuar processes coud be ofkey therapeutic vaue.

    Conclusions

    A number of processes resuting in AKI invove isch emia foowed by a compex interaction of various cetypes within the kidney. Epitheia ce injury mediatesfunctiona aterations through direct faiure of the ces totransport ions and moecues, or indirecty by mediating

    a decrease in GFR. Epitheia ces aso infuence the func tion of endotheia ces by reeasing chemokines, cyto kines, and other soube mediators. Interactions betweenendotheia ces and eukocytes contribute to continuedhypoxia, infammation, and further epitheia ce injuryand dysfunction. Numerous therapeutic targets havebeen identified that prevent or imit ongoing injury.Additiona approaches to improve repair and minimizefibrosis and vascuar dropout wi aso be critica in imit ing the deveopment of CKD and transition from CKDto end stage rena disease as a consequence of ischemicAKI in patients at high risk.

    Review criteria

    This Review was based on a search of the PubMed and

    OVID databases using a combination of search terms that

    included acute kidney injury, acute tubular necrosis,apoptosis, endothelial cell injury, inflammation,

    leukocytes, cytokines, coagulation, stem cells,

    endothelial progenitor cells, prerenal azotemia,

    cytoskeletal alterations, renal blood flow, glomerularfiltration rate, actin, oxidative stress, reactive

    oxygen species, heme oxygenase, heat shock

    proteins, repair, vascular permeability, neutrophils,

    T cells, and macrophages. Reference lists of selectedarticles were searched for further material. Articles were

    chosen based on their originality and relevance to this

    Review, and only English-language articles were selected.

    1. Thakar, C. V., Arrigain, S., Worley, S., Yared, J. P.& Paganini, E. P. A clinical score to predict acuterenal failure after cardiac surgery.J. Am. Soc.Nephrol.16, 162168 (2005).

    2. Singh, P., Rifkin, D. E. & Blantz, R. C. Chronic

    kidney disease: an inherent risk factor for acutekidney injury? Clin. J. Am. Soc. Nephrol.5,16901695 (2010).

    3. Coca, S. G. Acute kidney injury in elderlypersons.Am. J. Kidney Dis. 56, 122131(2010).

    4. Harel, Z. & Chan, C. T. Predicting and preventingacute kidney injury after cardiac surgery. Curr.Opin. Nephrol. Hypertens.17, 624628 (2008).

    5. James, M. T. et al. Glomerular filtration rate,proteinuria, and the incidence andconsequences of acute kidney injury: a cohortstudy. Lancet376, 20962103 (2010).

    6. Cruz, D. N., Bagshaw, S. M., Ronco, C. &Ricci, Z. Acute kidney injury: classification andstaging. Contrib. Nephrol.164, 2432 (2010).

    7. Ricci, Z., Cruz, D. & Ronco, C. The RIFLE criteria

    and mortality in acute kidney injury:a systematic review. Kidney Int. 73, 538546(2008).

    8. Molitoris, B. A., Melnikov, V. Y., Okusa, M. D. &Himmelfarb, J. Technology insight: biomarkerdevelopment in acute kidney injurywhat canwe anticipate? Nat. Clin. Pract. Nephrol.4,154165 (2008).

    9. Molitoris, B. A. Contrast nephropathy: are short-term outcome measures adequate forquantification of long-term renal risk? Nat. Clin.Pract. Nephrol.4, 594595 (2008).

    10. Himmelfarb, J. Acute kidney injury in the elderly:problems and prospects. Semin. Nephrol.29,658664 (2009).

    11. Liao, F. & Pascual, J. Epidemiology of acuterenal failure: a prospective, multicenter,

    community-based study. Madrid Acute RenalFailure Study Group. Kidney Int.50, 811818(1996).

    12. Nash, K., Hafeez, A. & Hou, S. Hospital-acquiredrenal insufficiency.Am. J. Kidney Dis.39,

    930936 (2002).13. Sesso, R., Roque, A., Vicioso, B. & Stella, S.Prognosis of ARF in hospitalized elderly patients.

    Am. J. Kidney Dis.44, 410419 (2004).14. Wencker, D. Acute cardio-renal syndrome:

    progression from congestive heart failure tocongestive kidney failure. Curr. Heart Fail. Rep.4,134138 (2007).

    15. Wan, L. et al. Pathophysiology of septic acutekidney injury: what do we really know? Crit. CareMed.36, S198S203 (2008).

    16. Himmelfarb, J. et al. Evaluation and initialmanagement of acute kidney injury. Clin. J. Am.Soc. Nephrol.3, 962967 (2008).

    17. Fujii, T. et al. The role of renal sympatheticnervous system in the pathogenesis of ischemicacute renal failure. Eur. J. Pharmacol. 481,

    241248 (2003).18. Blantz, R. C. The glomerular and tubular actionsof angiotensin II.Am. J. Kidney Dis. 10 (Suppl. 1),26 (1987).

    19. Kastner, P. R., Hall, J. E. & Guyton, A. C. Controlof glomerular filtration rate: role of intrarenallyformed angiotensin II.Am. J. Physiol.246,F897F906 (1984).

    20. Badr, K. F. & Ichikawa, I. Prerenal failure:a deleterious shift from renal compensation todecompensation.N. Engl. J. Med.319, 623629(1988).

    21. Maddox, D. & Brenner, B. M. in The Kidney6thedn Vol. 1 (eds Brenner, B. M. & Levine, S. A.)319374 (W. B. Saunders, Philadelphia, 2000).

    22. Yared, A., Kon, V. & Ichikawa, I. Mechanism ofpreservation of glomerular perfusion and

    filtration during acute extracellular fluid volumedepletion. Importance of intrarenal vasopressin-prostaglandin interaction for protecting kidneysfrom constrictor action of vasopressin.J. Clin.Invest.75, 14771487 (1985).

    23. Oliver, J. A., Sciacca, R. R. & Cannon, P. J. Renalvasodilation by converting enzyme inhibition.Role of renal prostaglandins. Hypertension5,166171 (1983).

    24. Cryer, H. G., Bloom, I. T., Unger, L. S. &Garrison, R. N. Factors affecting renalmicrovascular blood flow in rat hyperdynamicbacteremia.Am. J. Physiol.264, H1988H1997(1993).

    25. Molitoris, B. A. & Sutton, T. A. Endothelial injuryand dysfunction: role in the extension phase ofacute renal failure. Kidney Int.66, 496499(2004).

    26. Oberbauer, R., Rohrmoser, M., Regele, H.,Mhlbacher, F. & Mayer, G. Apoptosis of tubularepithelial cells in donor kidney biopsies predictsearly renal allograft function.J. Am. Soc. Nephrol.

    10, 20062013 (1999).27. Rosenberger, C. et al. Activation of hypoxia-inducible factors ameliorates hypoxic distaltubular injury in the isolated perfused rat kidney.Nephrol. Dial. Transplant.23, 34723478(2008).

    28. Alejandro, V. et al. Mechanisms of filtrationfailure during postischemic injury of the humankidney. A study of the reperfused renal allograft.

    J. Clin. Invest.95, 820831 (1995).29. Ramaswamy, D. et al. Maintenance and recovery

    stages of postischemic acute renal failure inhumans.Am. J. Physiol. Renal Physiol.282,F271F280 (2002).

    30. Solez, K., Morel-Maroger, L. & Sraer, J. D. Themorphology of acute tubular necrosis in man:analysis of 57 renal biopsies and a comparison

    REVIEWS

    2011 Macmillan Publishers Limited. All rights reserved

  • 8/3/2019 Fisiopatologia de IRA Isquemica

    11/12NATURE REVIEWS|NEPHROLOGY VOLUME 7 | APRIL 2011 | 199

    with the glycerol model. Medicine (Baltimore)58,362376 (1979).

    31. Racusen, L. inAcute Renal Failure 1st edn (edsMolitoris, B. A. & Finn, W. F.) 112(W. B. Saunders, Philadelphia, 2001).

    32. Saikumar, P. & Venkatachalam, M. A. Role ofapoptosis in hypoxic/ischemic damage in thekidney. Semin. Nephrol.23, 511521 (2003).

    33. Wagner, M. C. et al. Ischemic injury to kidneyinduces glomerular podocyte effacement and

    dissociation of slit diaphragm proteins Neph1and ZO-1.J. Biol. Chem.283, 3557935589(2008).

    34. Molitoris, B. A. Actin cytoskeleton in ischemicacute renal failure. Kidney Int.66, 871883(2004).

    35. Ashworth, S. L., Sandoval, R. M., Tanner, G. A. &Molitoris, B. A. Two-photon microscopy:visualization of kidney dynamics. Kidney Int.72,416421 (2007).

    36. Molitoris, B. A., Dahl, R. & Hosford, M. CellularATP depletion induces disruption of the spectrincytoskeletal network.Am. J. Physiol.271,F790F798 (1996).

    37. Ashworth, S. L., Sandoval, R. M., Hosford, M.,Bamburg, J. R. & Molitoris, B. A. Ischemic injuryinduces ADF relocalization to the apical domain

    of rat proximal tubule cells.Am. J. Physiol. RenalPhysiol.280, F886F894 (2001).38. Ashworth, S. L. et al. ADF/cofilin mediates actin

    cytoskeletal alterations in LLC-PK cells duringATP depletion.Am. J. Physiol. Renal Physiol.284,F852F862 (2003).

    39. Atkinson, S. J., Hosford, M. A. & Molitoris, B. A.Mechanism of actin polymerization in cellularATP depletion.J. Biol. Chem.279, 51945199(2004).

    40. Chen, J., Doctor, R. B. & Mandel, L. J.Cytoskeletal dissociation of ezrin during renalanoxia: role in microvillar injury.Am. J. Physiol.267, C784C795 (1994).

    41. Ashworth, S. L. et al. Renal ischemia inducestropomyosin dissociation-destabilizing microvillimicrofilaments.Am. J. Physiol. Renal Physiol.

    286, F988F996 (2004).42. Molitoris, B. A. & Marrs, J. The role of celladhesion molecules in ischemic acute renalfailure.Am. J. Med.106, 583592 (1999).

    43. Zuk, A., Bonventre, J. V., Brown, D. & Matlin, K. S.Polarity, integrin, and extracellular matrixdynamics in the postischemic rat kidney.Am. J.Physiol.275, C711C731 (1998).

    44. Molitoris, B. A., Geerdes, A. & McIntosh, J. R.Dissociation and redistribution of Na+, K+-ATPasefrom its surface membrane actin cytoskeletalcomplex during cellular ATP depletion.J. Clin.Invest.88, 462469 (1991).

    45. Molitoris, B. A. Na+-K+-ATPase that redistributesto apical membrane during ATP depletionremains functional.Am. J. Physiol.265,F693F697 (1993).

    46. Lieberthal, W., Koh, J. S. & Levine, J. S. Necrosisand apoptosis in acute renal failure. Semin.Nephrol.18, 505518 (1998).

    47. Bonegio, R. & Lieberthal, W. Role of apoptosis inthe pathogenesis of acute renal failure. Curr.Opin. Nephrol. Hypertens.11, 301308 (2002).

    48. Guo, R., Wang, Y., Minto, A. W., Quigg, R. J. &Cunningham, P. N. Acute renal failure inendotoxemia is dependent on caspaseactivation.J. Am. Soc. Nephrol.15, 30933102(2004).

    49. Safirstein, R. L. Acute renal failure: from renalphysiology to the renal transcriptome. Kidney Int.Suppl.91, S62S66 (2004).

    50. Nicholson, D. W. From bench to clinic withapoptosis-based therapeutic agents. Nature407, 810816 (2000).

    51. Edelstein, L. C., Lagos, L., Simmons, M.,Tirumalai, H. & Glinas, C. NF-B-dependentassembly of an enhanceosome-like complex onthe promoter region of apoptosis inhibitor Bfl-1/A1. Mol. Cell. Biol.23, 27492761 (2003).

    52. Kelly, K. J., Plotkin, Z., Vulgamott, S. L. &Dagher, P. C. P53 mediates the apoptoticresponse to GTP depletion after renal ischemia-reperfusion: protective role of a p53 inhibitor.

    J. Am. Soc. Nephrol.14, 128138 (2003).

    53. Park, K. M., Chen, A. & Bonventre, J. V.Prevention of kidney ischemia/reperfusion-induced functional injury and JNK, p38, andMAPK kinase activation by remote ischemicpretreatment.J. Biol. Chem.276, 1187011876(2001).

    54. Scheid, M. P., Schubert, K. M. & Duronio, V.Regulation of bad phosphorylation andassociation with Bcl-x

    Lby the MAPK/Erk kinase.

    J. Biol. Chem.274, 3110831113 (1999).55. Imamura, R. et al. Intravital 2-photon microscopy

    assessment of renal protection efficacy of siRNAfor p53 in experimental rat kidneytransplantation models. Cell Transplant.doi:10.3727/096368910X516619.

    56. Molitoris, B. A. et al. siRNA targeted to p53attenuates ischemic and cisplatin-induced acute

    kidney injury.J. Am. Soc. Nephrol.20,17541764 (2009).57. Sogabe, K. et al. Calcium dependence of

    integrity of the actin cytoskeleton of proximaltubule cell microvilli.Am. J. Physiol.271,F292F303 (1996).

    58. Portilla, D. Role of fatty acid beta-oxidation andcalcium-independent phospholipase A2 inischemic acute renal failure. Curr. Opin. Nephrol.Hypertens.8, 473477 (1999).

    59. Galli, F. et al. Oxidative stress and reactiveoxygen species. Contrib. Nephrol.149, 240260(2005).

    60. Li, L., Zepeda-Orozco, D., Black, R. & Lin, F.Autophagy is a component of epithelial cell fatein obstructive uropathy.Am. J. Pathol.176,17671778 (2010).

    61. Koesters, R. et al. Tubular overexpression oftransforming growth factor-1 inducesautophagy and fibrosis but not mesenchymaltransition of renal epithelial cells.Am. J. Pathol.177, 632643 (2010).

    62. Conger, J. D. & Schrier, R. W. Renalhemodynamics in acute renal failure.Annu. Rev.Physiol.42, 603614 (1980).

    63. Conger, J. D., Robinette, J. B. & Hammond, W. S.Differences in vascular reactivity in models ofischemic acute renal failure. Kidney Int.39,10871097 (1991).

    64. Noiri, E. et al. Oxidative and nitrosative stress inacute renal ischemia.Am. J. Physiol. RenalPhysiol.281, F948F957 (2001).

    65. Ling, H. et al. Attenuation of renal ischemia-reperfusion injury in inducible nitric oxide

    synthase knockout mice.Am. J. Physiol.277,F383F390 (1999).66. Goligorsky, M. S., Brodsky, S. V. & Noiri, E. NO

    bioavailability, endothelial dysfunction, andacute renal failure: new insights intopathophysiology. Semin. Nephrol.24, 316323(2004).

    67. Ogawa, T. et al. Contribution of nitric oxide to theprotective effects of ischemic preconditioning inischemia-reperfused rat kidneys.J. Lab. Clin.Med.138, 5058 (2001).

    68. Mattson, D. L. & Wu, F. Control of arterial bloodpressure and renal sodium excretion by nitricoxide synthase in the renal medulla.ActaPhysiol. Scand.168, 149154 (2000).

    69. Chander, V. & Chopra, K. Renal protective effectof molsidomine and L-arginine in ischemia-

    reperfusion induced injury in rats.J. Surg. Res.128, 132139 (2005).

    70. Sutton, T. A. et al. Injury of the renalmicrovascular endothelium alters barrierfunction after ischemia.Am. J. Physiol. RenalPhysiol.285, F191F198 (2003).

    71. Sutton, T. A. et al. Minocycline reduces renalmicrovascular leakage in a rat model of ischemicrenal injury.Am. J. Physiol. Renal Physiol.288,F91F97 (2005).

    72. Van de Wouwer, M., Collen, D. & Conway, E. M.Thrombomodulin-protein C-EPCR system:integrated to regulate coagulation andinflammation.Arterioscler. Thromb. Vasc. Biol.24,13741383 (2004).

    73. Gupta, A. et al. Activated protein C amelioratesLPS-induced acute kidney injury anddownregulates renal INOS and angiotensin 2.

    Am. J. Physiol. Renal Physiol.293, F245F254(2007).

    74. Gupta, A., Williams, M. D., Macias, W. L.,Molitoris, B. A. & Grinnell, B. W. Activatedprotein C and acute kidney injury: selectivetargeting of PAR-1. Curr. Drug Targets10,12121226 (2009).

    75. Mizutani, A., Okajima, K., Uchiba, M. &Noguchi, T. Activated protein C reduces

    ischemia/reperfusion-induced renal injury in ratsby inhibiting leukocyte activation. Blood95,37813787 (2000).

    76. Sharfuddin, A. A. et al. Soluble thrombomodulinprotects ischemic kidneys.J. Am. Soc. Nephrol.20, 524534 (2009).

    77. Tajra, L. C. et al.In vivo effects of monoclonalantibodies against rat

    2integrins on kidney

    ischemia-reperfusion injury.J. Surg. Res.87,3238 (1999).

    78. Singbartl, K., Forlow, S. B. & Ley, K. Platelet, butnot endothelial, P-selectin is critical forneutrophil-mediated acute postischemic renalfailure. FASEB J.15, 23372344 (2001).

    79. Burne, M. J. & Rabb, H. Pathophysiologicalcontributions of fucosyltransferases in renalischemia reperfusion injury.J. Immunol.169,

    26482652 (2002).80. Nemoto, T. et al. Small molecule selectin ligandinhibition improves outcome in ischemic acuterenal failure. Kidney Int.60, 22052214 (2001).

    81. Matsukawa, A. et al. Mice genetically lackingendothelial selectins are resistant to thelethality in septic peritonitis. Exp. Mol. Pathol.72,6876 (2002).

    82. Singbartl, K. & Ley, K. Leukocyte recruitment andacute renal failure.J. Mol. Med.82, 91101(2004).

    83. Basile, D. P. The endothelial cell in ischemicacute kidney injury: implications for acute andchronic function. Kidney Int.72, 151156(2007).

    84. Hrbelt, M. et al. Acute and chronicmicrovascular alterations in a mouse model of

    ischemic acute kidney injury.Am. J. Physiol.Renal Physiol.293, F688F695 (2007).85. Basile, D. P., Fredrich, K., Chelladurai, B.,

    Leonard, E. C. & Parrish, A. R. Renal ischemiareperfusion inhibits VEGF expression andinduces ADAMTS-1, a novel VEGF inhibitor.Am. J.Physiol. Renal Physiol.294, F928F936 (2008).

    86. Leonard, E. C., Friedrich, J. L. & Basile, D. P.VEGF-121 preserves renal microvessel structureand ameliorates secondary renal diseasefollowing acute kidney injury.Am. J. Physiol. RenalPhysiol.295, F1648F1657 (2008).

    87. Basi le, D. P. et al. Impaired endothelialproliferation and mesenchymal transitioncontribute to vascular rarefaction following acutekidney injury.Am. J. Physiol. Renal Physiol.doi:10.1152/ajprenal.00546.2010.

    FOCUS ON AKI IN CRIT ICAL CARE

    2011 Macmillan Publishers Limited. All rights reserved

  • 8/3/2019 Fisiopatologia de IRA Isquemica

    12/12

    88. Okusa, M. D., Chertow, G. M. & Portilla, D. forthe Acute Kidney Injury Advisory Group of theAmerican Society of Nephrology. The nexus ofacute kidney injury, chronic kidney disease, andWorld Kidney Day 2009. Clin. J. Am. Soc. Nephrol.4, 520522 (2009).

    89. Akcay, A., Nguyen, Q. & Edelstein, C. L.Mediators of inflammation in acute kidney injury.Mediators Inflamm.2009, 137072 (2009).

    90. Gluba, A. et al. The role of Toll-like receptors in

    renal diseases. Nat. Rev. Nephrol.6, 224235(2010).91. Wu, H. et al. TLR4 activation mediates kidney

    ischemia/reperfusion injury.J. Clin. Invest.117,28472859 (2007).

    92. Burne-Taney, M. J. & Rabb, H. The role ofadhesion molecules and T cells in ischemicrenal injury. Curr. Opin. Nephrol. Hypertens.12,8590 (2003).

    93. Burne-Taney, M. J. et al. B cell deficiency confersprotection from renal ischemia reperfusioninjury.J. Immunol. 171, 32103215 (2003).

    94. de Vries, B. et al. Complement factor C5amediates renal ischemia-reperfusion injuryindependent from neutrophils.J. Immunol. 170,38833889 (2003).

    95. Riedemann, N. C., Guo, R. F. & Ward, P. A. The

    enigma of sepsis.J. Clin. Invest. 112, 460467(2003).96. Huber-Lang, M. S. et al. Protective effects of

    anti-C5a peptide antibodies in experimentalsepsis. FASEB J. 15, 568570 (2001).

    97. Thurman, J. M. et al. C3a is required for theproduction of CXC chemokines by tubularepithelial cells after renal ishemia/reperfusion.

    J. Immunol. 178, 18191828 (2007).98. Thurman, J. M. et al. Altered renal tubular

    expression of the complement inhibitor Crrypermits complement activation after ischemia/reperfusion.J. Clin. Invest. 116, 357368(2006).

    99. Zheng, X. et al. Protection of renal ischemiainjury using combination gene silencing ofcomplement 3 and caspase 3 genes.

    Transplantation82, 17811786 (2006).100. Kinsey, G. R., Li, L. & Okusa, M. D. Inflammationin acute kidney injur y. Nephron Exp. Nephrol.109, e102e107 (2008).

    101. Tsuboi, N. et al. Roles of toll-like receptors inC-C chemokine production by renal tubularepithelial cells.J. Immunol. 169, 20262033(2002).

    102. El-Achkar, T. M. et al. Sepsis induces changes inthe expression and distribution of Toll-likereceptor 4 in the rat kidney.Am. J. Physiol. RenalPhysiol.290, F1034F1043 (2006).

    103. El-Achkar, T. M., Plotkin, Z., Marcic, B. &Dagher, P. C. Sepsis induces an increase in thickascending limb Cox-2 that is TLR4 dependent.

    Am. J. Physiol. Renal Physiol. 293, F1187F1196(2007).

    104. El-Achkar, T. M. et al. Tamm-Horsfall proteinprotects the kidney from ischemic injury bydecreasing inflammation and altering TLR4expression.Am. J. Physiol. Renal Physiol. 295,F534F544 (2008).

    105. Rusai, K. et al. Toll-like receptors 2 and 4 inrenal ischemia/reperfusion injury. Pediatr.Nephrol.25, 853860 (2010).

    106. Day, Y. J., Huang, L., Ye, H., Linden, J. &Okusa, M. D. Renal ischemia-reperfusion injuryand adenosine 2A receptor-mediated tissueprotection: role of macrophages.Am. J. Physiol.Renal Physiol.288, F722F731 (2005).

    107. Jo, S. K., Bajwa, A., Awad, A. S., Lynch, K. R. &Okusa, M. D. Sphingosine-1-phosphatereceptors: biology and therapeutic potential in

    kidney disease. Kidney Int.73, 12201230(2008).

    108. Bajwa, A. et al. Activation ofsphingosine-1-phosphate 1 receptor in theproximal tubule protects against ischemia-reperfusion injury.J. Am. Soc. Nephrol.21,955965 (2010).

    109. Dong, X. et al. Resident dendritic cells are thepredominant TNF-secreting cell in early renalischemia-reperfusion injury. Kidney Int.71,

    619628 (2007).110. Gandolfo, M. T. et al. Foxp3+ regulatory T cellsparticipate in repair of ischemic acute kidneyinjury. Kidney Int.76, 717729 (2009).

    111. Kinsey, G. R., Huang, L., Vergis, A. L., Li, L. &Okusa, M. D. Regulatory T cells contribute to theprotective effect of ischemic preconditioning inthe kidney. Kidney Int.77, 771780 (2010).

    112. Kinsey, G. R. et al. Regulatory T cells suppressinnate immunity in kidney ischemia-reperfusioninjury.J. Am. Soc. Nephrol.20, 17441753(2009).

    113. Li, L. et al. NKT cell activation mediatesneutrophil IFN- production and renal ischemia-reperfusion injury.J. Immunol.178, 58995911(2007).

    114. Gupta, A. et al. Distinct functions of activated

    protein C differentially attenuate acute kidneyinjury.J. Am. Soc. Nephrol.20, 267277 (2009).115. Liu, M. et al. Effect of T cells on vascular

    permeability in early ischemic acute kidney injuryin mice. Microvasc. Res.77, 340347 (2009).

    116. Savransky, V. et al. Role of the T-cell receptor inkidney ischemia-reperfusion injury. Kidney Int.69, 233238 (2006).

    117. Kelly, K. J. Distant effects of experimental renalischemia/reperfusion injury.J. Am. Soc. Nephrol.14, 15491558 (2003).

    118. Kramer, A. A. et al. Renal ischemia/reperfusionleads to macrophage-mediated increase inpulmonary vascular permeability. Kidney Int.55,23622367 (1999).

    119. Rabb, H. et al. Acute renal failure leads todysregulation of lung salt and water channels.

    Kidney Int.63, 600606 (2003).120. Liu, M. et al. Acute kidney injury leads toinflammation and functional changes in thebrain.J. Am. Soc. Nephrol.19, 13601370(2008).

    121. Pratschke, J. et al. Influence of donor brain deathon chronic rejection of renal transplants in rats.

    J. Am. Soc. Nephrol.12, 24742481 (2001).122. Pinsky, M. R. Pathophysiology of sepsis and

    multiple organ failure: pro- versus anti-inflammatory aspects. Contrib. Nephrol.144,3143 (2004).

    123. Kelly, K. J. Stress response proteins and renalischemia. Minerva Urol. Nefrol.54, 8191(2002).

    124. Paller, M. S., Weber, K. & Patten, M. Nitric oxide-mediated renal epithelial cell injury during

    hypoxia and reoxygenation. Ren. Fail.20,459469 (1998).125. Hill-Kapturczak, N., Chang, S. H. & Agarwal, A.

    Heme oxygenase and the kidney. DNA Cell Biol.21, 307321 (2002).

    126. Inguaggiato, P. et al. Cellular overexpression ofheme oxygenase-1 up-regulates p21 and confersresistance to apoptosis. Kidney Int.60,21812191 (2001).

    127. Kapturczak, M. H. et al. Heme oxygenase-1modulates early inflammatory responses:evidence from the heme oxygenase-1-deficientmouse.Am. J. Pathol.165, 10451053 (2004).

    128. Stromski, M. E. et al. Chemical and functionalcorrelates of postischemic renal ATP levels.Proc. Natl Acad. Sci. USA83, 61426145 (1986).

    129. Spiegel, D. M., Wilson, P. D. & Molitoris, B. A.Epithelial polarity following ischemia:a requirement for normal cell function.Am. J.Physiol.256, F430F436 (1989).

    130. Ichimura, T. et al. Kidney injury molecule-1 is aphosphatidylserine receptor that confers aphagocytic phenotype on epithelial cells.J. Clin.Invest.118, 16571668 (2008).

    131. Li, B. et al. The melanoma-associatedtransmembrane glycoprotein Gpnmb controls

    trafficking of cellular debris for degradation andis essential for tissue repair. FASEB J.24,47674781 (2010).

    132. Lin, S. L. et al. Macrophage Wnt7b is critical forkidney repair and regeneration. Proc. Natl Acad.Sci. USA107, 41944199 (2010).

    133. Matsumoto, M. et al. Induction ofrenoprotective gene expression by cobaltameliorates ischemic injury of the kidney inrats.J. Am. Soc. Nephrol. 14, 18251832(2003).

    134. Vannay, A. et al. Divergence of renal vascularendothelial growth factor mRNA expression andprotein level in post-ischaemic rat kidneys. Exp.Physiol.89, 435444 (2004).

    135. Ichimura, T. & Bonventre, J. V. inAcute RenalFailure 1st edn (eds Molitoris, B. A. & Finn, W. F.)

    101118 (W. B. Saunders, Philadelphia, 2001).136. Gupta, S. et al. Isolation and characterization ofkidney-derived stem cells.J. Am. Soc. Nephrol. 17, 30283040 (2006).

    137. Bussolati, B. et al. Isolation of renal progenitorcells from adult human kidney.Am. J. Pathol.166, 545555 (2005).

    138. De Broe, M. E. Tubular regeneration and therole of bone marrow cells: stem celltherapya panacea? Nephrol. Dial. Transplant.20, 23182320 (2005).

    139. Lange, C. et al. Administered mesenchymalstem cells enhance recovery from ischemia/reperfusion-induced acute renal failure in rats.Kidney Int.68, 16131617 (2005).

    140. Tgel, F., Zhang, P., Hu, Z. & Westenfelder, C.VEGF is a mediator of the renoprotective

    effects of multipotent marrow stromal cells inacute kidney injury.J. Cell. Mol. Med. 13,21092114 (2009).

    141. Humphreys, B. D. & Bonventre, J. V.Mesenchymal stem cells in acute kidney injury.

    Annu. Rev. Med. 59, 311325 (2008).142. Humphreys, B. D. et al. Intrinsic epithelial cells

    repair the kidney after injur y. Cell Stem Cell2,284291 (2008).

    143. Cantley, L. G. Adult stem cells in the repair ofthe injured renal tubule. Nat. Clin. Pract.Nephrol.1, 2232 (2005).

    144. Reinders, M. E., Rabelink, T. J. & Briscoe, D. M.Angiogenesis and endothelial cell repair inrenal disease and allograft rejection.J. Am. Soc.Nephrol. 17, 932942 (2006).

    145. Tongers, J. & Losordo, D. W. Frontiers in

    nephrology: the evolving therapeuticapplications of endothelial progenitor cells.J. Am. Soc. Nephrol. 18, 28432852 (2007).

    146. Becherucci, F. et al. The role of endothelialprogenitor cells in acute kidney injur y. BloodPurif.27, 261270 (2009).

    147. Li, B. et al. Mobilized human hematopoieticstem/progenitor cells promote kidney repairafter ischemia/reperfusion injury. Circulation121, 22112220 (2010).

    Author contributions

    A. A. Sharfuddin and B. A. Molitoris contributedequally to researching data for the article, discussionof the content, writing and reviewing/editing of themanuscript before submission.

    REVIEWS