17
1. Introduction 2. Current and new animal models of AKI 3. Need for new animal models of AKI 4. Advances in AKI animals models 5. New potential therapeutic approaches to AKI characterized in experimental models 6. Clinical therapies for AKI treatment 7. Conclusions 8. Expert opinion Review Progress in the development of animal models of acute kidney injury and its impact on drug discovery Ana B Sanz, Marı ´a Dolores Sanchez-Nin ˜o, Catalina Martı ´n-Cleary, Alberto Ortiz & Adria ´n M Ramos Fundacio´n Renal In˜igo A ´ lvarez de Toledo (FRIAT), Madrid, Spain Introduction: Acute kidney injury (AKI) is a clinical syndrome characterized by the acute loss of kidney function. AKI is increasingly frequent and is associated with impaired survival and chronic kidney disease progression. Experimental AKI models have contributed to a better understanding of pathophysiological mech- anisms but they have not yet resulted in routine clinical application of novel therapeutic approaches. Areas covered: The authors present the advances in experimental AKI models over the last decade. Furthermore, the authors review their current and expected impact on novel drug discovery. Expert opinion: New AKI models have been developed in rodents and non- rodents. Non-rodents allow the evaluation of specific aspects of AKI in both bigger animals and simpler organisms such as drosophila and zebrafish. New rodent models have recently reproduced described clinical entities, such as aristolochic and warfarin nephropathies, and have also provided better mod- els for old entities such as thrombotic microangiopathy-induced AKI. Several therapies identified in animal models are now undergoing clinical trials in human AKI, including p53 RNAi and bone-marrow derived mesenchymal stem cells. It is conceivable that further refinement of animal models in com- bination with ongoing trials and novel trials based on already identified potential targets will eventually yield effective therapies for clinical AKI. Keywords: apoptosis, fibrosis, inflammation, kidney, necroptosis, nephrotoxicity, transgenic mice, TWEAK Expert Opin. Drug Discov. [Early Online] 1. Introduction Acute kidney injury (AKI) is a clinical syndrome characterized by the acute loss of kidney function that leads to increased serum creatinine or oliguria [1]. AKI may result in the need for renal replacement therapy (RRT). However, even when not needing RRT, AKI is still a serious disorder, conferring an increased risk of death that persists for over a year following the AKI episode and also increasing the risk of chronic kidney disease (CKD) progression [2-5]. AKI and CKD have been recently recognized as an integrated clinical syndrome and much research is being carried out on the interface of both clinical conditions [6]. Indeed, AKI and CKD share key bio- logical processes such as cell death, cell proliferation, cell dedifferentiation, inflam- mation and fibrosis and also share biomarkers [7-10]. In general, the magnitude of these processes is greater in AKI than in CKD, except for fibrosis which is associated to chronicity. The short time-course and the severity of histological and functional changes have fueled the study of experimental animal models of AKI to characterize mediators involved in kidney injury in order to identify novel therapeutic 10.1517/17460441.2013.793667 © 2013 Informa UK, Ltd. ISSN 1746-0441, e-ISSN 1746-045X 1 All rights reserved: reproduction in whole or in part not permitted Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sydney on 05/13/13 For personal use only.

Progress in the development of animal models of acute kidney injury and its impact on drug discovery

Embed Size (px)

Citation preview

Page 1: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

1. Introduction

2. Current and new animal

models of AKI

3. Need for new animal models of

AKI

4. Advances in AKI animals

models

5. New potential therapeutic

approaches to AKI

characterized in experimental

models

6. Clinical therapies for AKI

treatment

7. Conclusions

8. Expert opinion

Review

Progress in the development ofanimal models of acute kidneyinjury and its impact on drugdiscoveryAna B Sanz, Marıa Dolores Sanchez-Nino, Catalina Martın-Cleary,Alberto Ortiz† & Adrian M Ramos†Fundacion Renal Inigo Alvarez de Toledo (FRIAT), Madrid, Spain

Introduction: Acute kidney injury (AKI) is a clinical syndrome characterized

by the acute loss of kidney function. AKI is increasingly frequent and is associated

with impaired survival and chronic kidney disease progression. Experimental AKI

models have contributed to a better understanding of pathophysiological mech-

anisms but they have not yet resulted in routine clinical application of novel

therapeutic approaches.

Areas covered: The authors present the advances in experimental AKI models

over the last decade. Furthermore, the authors review their current and

expected impact on novel drug discovery.

Expert opinion: New AKI models have been developed in rodents and non-

rodents. Non-rodents allow the evaluation of specific aspects of AKI in both

bigger animals and simpler organisms such as drosophila and zebrafish. New

rodent models have recently reproduced described clinical entities, such as

aristolochic and warfarin nephropathies, and have also provided better mod-

els for old entities such as thrombotic microangiopathy-induced AKI. Several

therapies identified in animal models are now undergoing clinical trials in

human AKI, including p53 RNAi and bone-marrow derived mesenchymal

stem cells. It is conceivable that further refinement of animal models in com-

bination with ongoing trials and novel trials based on already identified

potential targets will eventually yield effective therapies for clinical AKI.

Keywords: apoptosis, fibrosis, inflammation, kidney, necroptosis, nephrotoxicity, transgenic

mice, TWEAK

Expert Opin. Drug Discov. [Early Online]

1. Introduction

Acute kidney injury (AKI) is a clinical syndrome characterized by the acute loss ofkidney function that leads to increased serum creatinine or oliguria [1]. AKI mayresult in the need for renal replacement therapy (RRT). However, even when notneeding RRT, AKI is still a serious disorder, conferring an increased risk of deaththat persists for over a year following the AKI episode and also increasing the riskof chronic kidney disease (CKD) progression [2-5]. AKI and CKD have been recentlyrecognized as an integrated clinical syndrome and much research is being carried outon the interface of both clinical conditions [6]. Indeed, AKI and CKD share key bio-logical processes such as cell death, cell proliferation, cell dedifferentiation, inflam-mation and fibrosis and also share biomarkers [7-10]. In general, the magnitude ofthese processes is greater in AKI than in CKD, except for fibrosis which is associatedto chronicity. The short time-course and the severity of histological and functionalchanges have fueled the study of experimental animal models of AKI to characterizemediators involved in kidney injury in order to identify novel therapeutic

10.1517/17460441.2013.793667 © 2013 Informa UK, Ltd. ISSN 1746-0441, e-ISSN 1746-045X 1All rights reserved: reproduction in whole or in part not permitted

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 2: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

approaches and biomarkers. However, despite many success-ful reports in animal models, no novel biomarker resultingfrom these studies is currently in use in routine clinical prac-tice, and there is no routine therapeutic approach to AKIbased on experimental advances on the molecular and cellularmechanisms of kidney injury. This lack of tools to preventinjury or accelerate recovery reflects a still incomplete under-standing of molecular and cellular events, the deficiencies ofcurrently available animal models and probably a lack of accu-rate tools for clinical staging of AKI. In this regard, the cur-rent clinical definition of AKI based on functional criteriaidentifies late events in the course of kidney injury. A noveldefinition that allows an earlier diagnosis and therapeuticintervention is required and a search for novel biomarkers isunderway. Animal models of AKI will play a decisive role inthe quest for biomarkers and novel therapeutic strategies.We now review current and upcoming animal models ofAKI, illustrate some emerging therapeutic approaches identi-fied in recent years by the use of these models, critically reviewongoing and recent clinical trials based on experimentalmodel data and provide expert opinion on the current stateof the field and unmet medical needs.

2. Current and new animal models of AKI

AKI is a syndrome that recognizes many triggering events inclinical situations, including nephrotoxic therapeutic drugs,environmental contaminants and poisons, urinary tractobstructions, bacterial toxins, ischemic episodes and, fre-quently in clinical practice, a sequential or concomitant pres-ence of several of these factors. Thus, several experimentalanimal models have been developed to mimic these differentpotential clinical settings (Table 1). Comprehensive reviewsof the available models, their advantages and limitations,

were performed more than a decade ago [11]. More recentlytechnical details for a wide range of ischemia-reperfusion,toxic and septic models have been collected [12]. We referthe readers to these manuscripts for an in-depth descriptionof traditional models of AKI, while we will concentrate, inthe present review, on emerging models of AKI and theimpact of experimental models on drug discovery.

3. Need for new animal models of AKI

Animal models of AKI have proved to be valuable in under-standing molecular mechanisms underlying AKI initiationand progression, but they also have limitations that haveprecluded widespread clinical translation. Thus, AKIremains an entity devoid of specific therapy, beyond symp-tomatic therapy, and all clinical trials completed to datebased on preclinical data have failed. Several factors contrib-ute to this failure including animal models that do not fullyreproduce the clinical situation, such as the classic ischemia/reperfusion model, relative timing of insult and therapy(most successful preclinical interventional approaches areprophylactic), the insufficient biomarker resources inhumans to stage the disease and, potentially, interspeciesdifferences. The translational gap between experimentaltherapies for AKI and their effective application in the clin-ical setting remains a concern in the scientific community.In order to overcome drawbacks detracting reliability fromusually employed in vivo models for AKI, researchers con-tinue to develop novel models that try to best fit clinicalconditions or better dissect the role of different moleculesor cells or allow a more detailed follow-up of molecular orcellular events. New proposed in vivo models focus on tar-geting specific kidney areas and epithelial subtypes as wellas pathological events previously identified as the startingpoint to a further widespread renal damage, a purpose diffi-cult to achieve with traditional models. Many new animalmodels deal with genetically modified mice. These animalsmay express proteins that under specific conditions damagethe cellular type where they are expressed, explore the roleplayed by specific molecules, molecular pathways and bio-chemical processes underlying experimental AKI in tradi-tional models or are designed to closely follow-up cell fateor molecular events (Table 1).

4. Advances in AKI animals models

Recent advances in AKI animal models have focused onimproving previous models or developing new models tomeet previously unmet needs usually in rodent animal modelsbut sometimes expanded into non-rodents or employinggenetically modified animals. Specific attention has beendevoted to address the AKI to CKD transition and to repro-duce special high-risk clinical situations that predispose toAKI initiation and progression.

Article highlights.

. Experimental AKI models have contributed to a betterunderstanding of pathophysiological mechanisms buthave not resulted yet in routine clinical therapy for AKI.

. New AKI models have been developed in rodents andnon-rodents.

. Non-rodents allow the evaluation of specific aspects ofAKI in both bigger animals and simpler organisms.

. New rodent models have reproduced recently describedclinical entities, have provided better models for oldentities and have allowed exploration of factorspredisposing to AKI.

. Genetically modified animals allow the functionalevaluation of specific molecules or tubular segments,lineage tracing and real-time monitoring oftubular injury.

. In recent years a spate of novel druggable targets hasbeen identified and clinical trials are underway for someof them.

This box summarizes key points contained in the article.

A. Sanz et al.

2 Expert Opin. Drug Discov. [Early Online]

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 3: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

Table 1. Traditional and novel AKI animal models.

Model Characteristics

RodentsToxic or drug-induced AKI*Therapeutic drugs:

Cisplatin [11,12]

Aminoglycosides [11,12]

Nonsteroidal anti-inflammatory drugs [128]

Acetaminophen (Paracetamol) [12]5/6 nephrectomy + warfarin [30]

Non-therapeutic drugs:Folic acid [12]

z

Diagnostic drugs:Radiocontrast [12]

Poisons/ambient contaminants:Ethylene glycol [140]z

Aristolochic acid [129]

Mercury compounds and derivatives [36]

Pigment-inducedGlycerol-induced rhabdomyolysis [12]

OtherMaleate nephrotoxicity [23]

AdvantagesSimplicity and reproducibilityClinical correlate: may simulate renal adverse effects of pharmacologicaltreatments and common intoxications or poisoning affecting the kidneyShort time to AKI production (1 -- 6 days) and achieved with single orrepetitive dosePredisposing factors for human AKI can be reproduced in animal modelsEvolution of damage and renal function, including the recovery phase, parallelhuman AKIUseful to study long-term consequences of AKI, including transition to CKDand fibrosis in some modelsCan be performed in small animals

DisadvantagesDose to produce AKI can be much higher than those for human use, raisingquestions of clinical significanceWidespread kidney cell damage beyond tubular cellsNo adequate mouse models of radiocontrast-induced AKINo clinical correlate of maleate nephrotoxicity, single case report of accidentalfolic acid nephrotoxicity

Observations5/6 nephrectomy + warfarin is the only model available to study AKI caused byanticoagulants. Only available in ratsThere are alternative methods to study rhabdomyolysis including myoglobin orhemoglobin infusion but they poorly reproduce human physiopathologicalaspectsMaleate causes proximal tubular cell specific coenzyme A depletion, ATP andGSH depletion simulating ischemiaAristolochic acid induces rapidly progressive CKD and cancer in aristolochicacid nephropathy and has been implicated in Balkan nephropathy, allowing theexploration of the AKI/CKD/cancer interface

Obstructive AKI§

UUO [11]

Adenine ingestion [130]

Cast nephropathy following immunoglobulinlight chain injection [112]

Folic acid [12]z

Ethylene glycol [87]z

AdvantagesUUO: accelerated model of AKI leading to renal fibrosisAllows variations in the timing, severity and duration. Reversal of theobstruction permits the study of recovery. It presents with tubular damage(apoptosis and necrosis) (UUO)Technically simple and reproducible (adenine ingestion, cast nephropathy)Clinical relevance of UUO and cast nephropathy, the latter in the setting ofoverproduction of monoclonal immunoglobulin free light chains, clinicallyassociated to multiple myelomaCan be performed in small animals

DisadvantagesUUO requires surgery. Not widely used as AKI modelOwing to their novelty, adenine ingestion and cast nephropathy need furthervalidationUUO: renal function cannot be measured since it is compensated by the non-ligated kidney

Ischemia-reperfusion injury (IRI)§

Time-dependent ischemia [11]

Warm/cold [11,12]

AdvantagesClinically very relevant, including for kidney transplantsThe most employed model to study AKI which give a high knowledgebackground

*A more comprehensive list is provided in Refs [11] and [12].zBoth toxicity and intratubular precipitation contribute to AKI.§Additional details in reference to these models are given in Ref. [11].{Experimental AKI has been performed in genetically modified mice in order to explore the role of specific molecules in AKI. Here, we reflect examples of the use

of genetically modified mice to answer more general questions about AKI, such as the role of specific cell types, the sensitization to a particular form of kidney

injury or cell tagging for better assessment of specific aspects of AKI.

Progress in the development of animal models of AKI and its impact on drug discovery

Expert Opin. Drug Discov. [Early Online] 3

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 4: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

Table 1. Traditional and novel AKI animal models (continued).

Model Characteristics

Unilateral or bilateral [11,12]Renal artery occlusion + E. coli injection [131]

The variable ischemia time allows different forms of injuryCan be performed in small animals

DisadvantagesMurine model is less reproducible than the rat modelTechnical and surgical procedures require carefully monitoring and surgeonsmust be well trained. Close post-surgery care is also neededMost common clinical situation is prolonged low level ischemia with potentialrepeat episodes, while the experimental setting implies complete short-term occlusion

ObservationsBilateral ischemia is more relevant to human pathology where abnormal bloodsupply is usually bilateral

Endotoxic AKI§

LPS injection or infusion [11]

Cecal puncture/ligation [11]

Bacterial infusion [11]

Uterine obstruction + bacterial inoculation [72]

AdvantagesLPS injection is simple and inexpensiveThe dose of endotoxin and bacteria can be standardizedLPS mimic systemic inflammatory reaction occurring in sepsisLeaking of cecal bacteria avoid transient inflammatory response of toxininjectionAppropriate for small animals

DisadvantagesVariable hemodynamic response between modelsMultiorgan dysfunction is dependent on the modelThe extent of renal damage is variable and the acute tubular necrosis is notalways achievedThere is a variable outcome with bacterial strainSurgical methods are not well standardizedLPS does not reproduce the presence of live bacteria occurring in the clinicalsituationCecal puncture/ligation involves surgery. Use of antibiotics in the clinicalsituation differs from animal model

Thrombotic microangiopathy-induced AKIStx injection to different strains of mice [26]

LPS + Stx [29]

Genetically modified Stx-expressingbacteria [27]

AdvantagesClinically relevant for HUS, the most common cause of AKI in childrenStx injection simple and inexpensiveThe dose of toxin and bacteria can be standardizedGenetically modified Stx-expressing bacteria allow reproduction of theinfectious agent-driven clinical situationAppropriate for small animals

DisadvantagesStx does not reproduce the presence of live bacteria occurring in the clinicalsituation

ObservationsCAST/Ei mice are more susceptible to thrombotic diseases than C57BL/6

Selected examples of genetically modified mice{

Ischemia-reperfusion in PKD KO mice [132] Allows study of mechanisms of cystogenesisSelective expression of human HB-EGF (thediphtheria toxin receptor, hDTR) to sensitize todiphtheria toxin [48,49]

Selective expression of HSV1-tk transgene tosensitize to ganciclovir [52]

Allows selective killing of cells expressing these proteins

Genetically labeled cells [52] Allows lineage tracingNgal reporter mice [55] Allows in vivo monitoring and localization of kidney injury

*A more comprehensive list is provided in Refs [11] and [12].zBoth toxicity and intratubular precipitation contribute to AKI.§Additional details in reference to these models are given in Ref. [11].{Experimental AKI has been performed in genetically modified mice in order to explore the role of specific molecules in AKI. Here, we reflect examples of the use

of genetically modified mice to answer more general questions about AKI, such as the role of specific cell types, the sensitization to a particular form of kidney

injury or cell tagging for better assessment of specific aspects of AKI.

A. Sanz et al.

4 Expert Opin. Drug Discov. [Early Online]

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 5: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

4.1 Novel rodent AKI modelsMouse and rat rodent models are by far the most used tostudy AKI pathological events, potential druggable targetsand select specific biomarkers of disease. Mice and rats arerelatively inexpensive to grow and maintain, they are fastbreeders and their genetics and physiology have been widelystudied and can be easily compared to humans. Indeed,mice, rats and humans share 99% of genes [13,14]. Rats arelarger than mice and this may be an advantage for techni-cally demanding models. However, mice constitute anextraordinary platform to get genetically engineered animalscarrying induced mutations and there are more gene tar-geted mice than rats. However, tools currently available tocreate genetically modified mice are not applicable to rats.Instead, genetically modified rats can be obtained frompoint mutations induced in master genes by treating maleswith usual chemical mutagens and further mating tountreated females, then followed by selection of disease phe-notypes of interest carrying a traceable mutation. In addi-tion, other genetic tools are currently being developed [15].Recently, a novel approach based on chromosome substitu-tion illustrates the way that genetic approaches in the ratmay help to identify protective genes against AKI. Genesexpressed in intrinsic AKI-resistant strains were transferredto other AKIs susceptible to create a consomic strain, thusconferring this later with the same natural component ofresistance [16].

Novel rodent AKI models have been developed in the pastdecade that address novel clinical entities or try to improvemodeling of clinical conditions. These add to already availableAKI models (Table 1). We will now briefly review some selectedexamples of experimental AKI induced by specific exogenoustoxins, thrombotic microangiopathy and warfarin-inducedhematuria that try to more closely reproduce relatively novelclinical situations (aristolochic acid or warfarin nephropathies)or clinical situations requiring improved modeling (thromboticmicroangiopathy and ischemia-reperfusion injury).

4.1.1 Exogenous toxinsAristolochic acid was long known to induce AKI in rats andmice [17]. However, it was not until the 1994 description ofChinese herbs nephropathy (hereafter named as aristolochicacid nephropathy to avoid referring in totality all Chinese herbsas causal of nephropathy), a rapidly progressive interstitial renalfibrosis initially reported in young women on a slimming regi-men including Chinese herbs, that interest in understanding thepathophysiogical mechanism of kidney injury boomed [18]. Ini-tially, rabbit and rat models focused on reproducing the humanCKD and helped to establish a link between this toxin and Bal-kan endemic nephropathy [19-21], but more recent mice modelshave been successfully used to identify new druggable targets inAKI and its transition to CKD, such as Jun N-terminal kinases(JNK) signaling role in G2/M arrest during AKI leading toresidual fibrosis [22].

Table 1. Traditional and novel AKI animal models (continued).

Model Characteristics

Whole mouse or cell specific KO oroverexpression of potential mediators of AKI

Allows characterization of specific molecular pathway or functionsAllows functional exploration of gene products in vivoAdvantagesThe main advantages have been specified for each model

DisadvantagesGenetically engineered mice are technically laborious to obtain and requirespecific knowledge on geneticsMay not be freely available

Non-rodentPigs and AKI involving surgical procedures,including kidney transplantation andcardiovascular surgery [83,133]

AdvantagesSize and physiology closer to humanMay one day be a source of xenografts

DisadvantagesBig size requires complex logistics and breeding facilities

Drosophila: malpighian tubules and adult kidneystem cells [92]

Zebrafish: gentamicin and cisplatinnephrotoxicity [91]

AdvantagesLow technical demands for breedingPossibility of high-throughput chemical or mutagenesis screensZebrafish kidneys contain self-renewing nephron stem/progenitor cells: studymolecular pathways for reactivation of this program in mammals [93]

DisadvantagesSimpler kidneys with less physiological similarity to humans

*A more comprehensive list is provided in Refs [11] and [12].zBoth toxicity and intratubular precipitation contribute to AKI.§Additional details in reference to these models are given in Ref. [11].{Experimental AKI has been performed in genetically modified mice in order to explore the role of specific molecules in AKI. Here, we reflect examples of the use

of genetically modified mice to answer more general questions about AKI, such as the role of specific cell types, the sensitization to a particular form of kidney

injury or cell tagging for better assessment of specific aspects of AKI.

Progress in the development of animal models of AKI and its impact on drug discovery

Expert Opin. Drug Discov. [Early Online] 5

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 6: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

Table

2.Someexamplesofmolecu

lartargets

exploredin

recentexperimentalAKImodels.

Candidate

target

Anim

almodel

Species

Targetingprocedure

Phenotypic

resp

onses

Renalfunction

Refs.

JNK,p53

UUO,IRI,aristolochic

acid

Mouse

PIF-a

(p53inhibitor)

SP600125(pan-JNKinhibitor)

#G2/M

arrest

#Fibrosis

Notassessed

[22]

CXCR4,CXCR7

Stx-m

ediated

Mouse

CXCR4antagonist

(AMD3100/plerixafor)

"Survival

Inpart

restoredBUN,sC

r[26]

Oat1

Mercury(II)chloride

Rat

Mouse

Oat1-KO

mice

#Histopathologicaldamage

NotchangeBUN

[36]

Smad3

IRI

Mouse

Smad-/-mice

#Histopathologicalinjury,renalIL-6

andendothelin,bloodIL-6

#BUN,sC

r[37]

MMP9

FA,IRI

Mouse

MPP9-/-mice

"Tubulardilationandapoptosis

Delayedrecovery

ofsC

r[38]

Dicer

IRI

Mouse

Dicer(flox/flox) X

CREY(PT-Dicer-/-)mice

#Tissuedamageandapoptosis

"Survival

#BUN,sC

r[39]

CCR1

IRI

Mouse

CCR1antagonism

(BX471)

CCR1-/-mice

#Leukocyte

infiltration#r

enal

CCL3,CCL5

NotchangesC

r[40]

Tweak

FAMouse

TweakKO

mice

#Tubularapoptosisandproliferation

#sCr

[41]

Fn14

IRI

Mouse

Fn14blockade(ITEM-2

blockingantibody)

#Inflammation

#Apoptosisandfibrosis

#BUN,sC

r[42]

ATF3

IRI

Mouse

ATF3-KO

mice

"Inflammation,apoptosis

"BUN,sC

r[43]

ATF3

genetransfer

#Apoptosis

"Survival

#BUN,sC

r

HO-1

Glycerol,cisplatin

Mouse

HO-1

transgenic

"Survival

#sCr

[44]

CSF-1

IRIDT-induced

Mouse

GW2580(c-fmsinhibitor)

Csf1-/-mice

Persistentrenalinjury

"BUN

[48]

TGFb

/Smadpathway

IRI

Mouse

THR-123(Alk3peptideagonist)

BMP7

#Tubulardamage

#Fibrosis

NochangeBUN

[66]

Dnmt1/Rasal1

pathway

FAMouse

5-Azacytidine

Dnmt1

+/-mice

#Progressionto

fibrosis

#sCr

[67]

Renalprogenitors

cells

(RMPs)

expressingCXCR7/CXCR4

Glycerol

Mouse

RMPswith/w

ithoutanti-CXCR4,

anti-CXCR7antibodies

CXCR4antagonist(AMD3100)

#Severity,fibrosis

(CXCR4orCXCR7antagonism

prevents

beneficialeffects)

Recovered

[110]

BUN:Bloodureanitrogen,FA

:Folic

acid,IRI:Ischemia-reperfusioninjury,sC

r:Serum

creatinine,UUO:Unilateralureteralobstruction.

A. Sanz et al.

6 Expert Opin. Drug Discov. [Early Online]

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 7: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

Maleate-induced proximal tubular cell toxicity and AKI inmice had received little attention because of a lack of a clinicalcorrelate. However, recent studies have ‘rediscovered’ thisform of renal injury as having similarities to metabolic eventsin ischemic/hypoxic tubular cell death, such as proximal tubu-lar cell specific coenzyme A, ATP and glutathione (GSH)depletion-simulating ischemia [23].

4.1.2 Thrombotic microangiopathyThere have also been advances in thrombotic microangiop-athy-induced AKI, as can be observed during human hemo-lytic uremic syndrome (HUS), the most common cause ofAKI in children. This is a rare disease in need of adequate ani-mal models and although great advances have been made inthe pathogenesis and treatment with eculizumab of atypical,genetic complement defect-related HUS [24], there is stillmuch confusion about therapy of typical HUS, as evidencedby the recent 2011 German outbreak where none of the tradi-tional forms of therapy (plasmapheresis and corticosteroids)proved beneficial and the long-discarded antibiotic therapyappeared to have a role [25]. An adequate choice of mousestrain and specific Shiga toxin (Stx) appears to be important.Shiga toxin 2 (Stx2) injection to CAST/Ei mice reproducedthe disease better than C57BL/6 mice, which are less suscep-tible to thrombotic diseases [26]. In CAST/Ei mice, a singlei.v. dose-dependent injection of Stx2 resulted in increasedmortality and, after 4 days, AKI with severe defects in corticalperfusion of the functional vasculature and increased urinealbumin/creatinine ratios [26]. Stx2 is the isoform associatedwith the most severe forms of human disease. The 2011German outbreak was caused by Stx2-producing Escherichiacoli. However, toxin injection does not reproduce the infec-tious nature of typical human HUS, usually the result ofenteric infection by toxin-expressing enterohemorrhagicE. coli (EHEC). However, EHEC infection in conventionalmice does not manifest key features of the disease, such asattaching and effacing lesions, intestinal damage and systemicillness. Genetic modification of the murine pathogen Citro-bacter rodentium has been used to express Stx-reproducedintestinal epithelium and Stx-dependent intestinal inflamma-tory damage and kidney damage [27]. Additional modelsinclude murine AKI induced by infusion of concanavalin Aand anti-concanavalin A antibodies [28] and lipopolysaccha-ride (LPS) plus Stx administration [29]. A transgenic murinemodel of atypical HUS is described in the next section.

4.1.3 Warfarin nephropathyA new rat model of warfarin-induced hematuric AKI couldonly be reproduced in five of six nephrectomized animalswith prior CKD [30]. This model provided a biological basisfor the recently described warfarin nephropathy syndrome ofAKI in humans overcoagulated with oral agents and will allowthe development of therapeutic approaches [31]. In this regard,hematuria results in kidney recruitment of macrophagesexpressing the CD163 hemoglobin scavenger receptor, and

hemoglobin toxicity is probably the key element in macro-scopic hematuria-associated AKI in IgA nephropathy [32-35].The need for prior five of six nephrectomies makes thebigger-sized rat a better animal model than the mouse andunderscores the clinical observation that prior CKD is a riskfactor for this form of AKI [31].

In summary, the AKI researcher is presented with anexpanding choice of potential models. Potential risk factors(e.g., diabetes and aging) in the targeted clinical entity aswell as method for inducing AKI (toxin, drug, route, dosing,animal strain and genomic background) are all aspects to beconsidered when choosing a model to identify and test drugcandidates.

4.2 Genetically modified miceMice are well suited for genetic modification involving tar-geted knockout (KO) or knock-in genes or able to condition-ally express proteins of interest. A growing body of AKIresearch is carried out in genetically modified mice, exploitingsome of these possibilities. Genetically modified mice havebeen used to perform functional studies of the role of specificgenes in AKI, to induce targeted injury of specific tubular seg-ments, for lineage tracing and for imaging of injured tubularsegments.

4.2.1 Functional studies of the role of specific genesGenetically modified mice have been used to address the func-tional role of specific AKI mediators (Table 2). Approximately15% of the mouse genome genes have been successfully targetedby means of genetic engineering tools. As the number of poten-tial AKI mediators amply exceeds those studied in geneticallymodified mice, further use of this technology in drug discoveryis expected. Already studied KO mice include, but are notlimited to, renal transporters (Oat1/Slc22a6), molecular effec-tors of relevant intracellular pathways (Smad3), metalloprotei-nases (MMP9), RNA regulators (Dicer), cytokines orreceptors (CCR1, TWEAK and Fn14) and transcription factors(ATF3) [36-43]. KO mice have been subjected to different mod-els of AKI to evaluate modification of the injury as a result ofthe gene manipulation and, thus, the role of the targeted genein AKI. These modifiers were manipulated, for example, inmercury- [36], folic acid- [38,41] and ischemia-induced AKI back-grounds [37,39,40,43]. Humanized transgenic mice were used toevaluate beneficial HO-1 induction during AKI [44]. TransgenicCfh-/-.FHD16-20 mice express a genetically modified comple-ment H protein that mimics the human mutations reportedin atypical HUS [45]. These animals develop spontaneous micro-angiopathy and AKI, thus providing a model to investigate therole of C5 complement factor in atypical HUS.

4.2.2 Targeted injury of specific tubular segmentsThe kidney is a complex organ composed of several epitheliaand other metabolically active cell types that interact withone another. Tubular epithelial cells are considered key targetsin AKI and may undergo death, dedifferentiaton and

Progress in the development of animal models of AKI and its impact on drug discovery

Expert Opin. Drug Discov. [Early Online] 7

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 8: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

activation [9]. However, most classic models of AKI are notselective and promote diffuse tubular cell injury, frequentlyin several tubular types, in addition to vascular and glomeru-lar injury; the relative role of tubular cell toxicity should beclarified. Proximal tubules are the main targets of injury forseveral nephrotoxins, including tenofovir and cidofovir thataccumulate into these cells because of their particular set oftransporters [46,47]. These transporters may be used in target-ing therapeutic agents to proximal tubules as a strategy totreat AKI.

4.2.2.1 Targeted injury of proximal tubulesSeveral genetically modified systems have been used foraddressing the specific role of proximal tubules in AKI. Trans-genic mice are used to induce selective tubular lesions and tospecifically study how injury primarily affects specific tubularcells and how this lesion expands to other cell types. Takingadvantage of the fact that rodents are 103 -- 106 times moreresistant to diphtheria toxin than other mammals, becausethe toxin does not bind to their heparin-binding epidermalgrowth factor (HB-EGF), selective expression of humanHB-EGF (the diphtheria toxin receptor, hDTR) in cells ofinterest allows induction of targeted injury. Genetically mod-ified mice that express hDTR under diverse promoters allowsensitization of proximal tubular cells, S3 segment proximaltubular cells or kidney epithelial cells to diphtheria toxinallowing selective killing of these cells upon diphtheria toxinadministration [48-50]. In mice expressing hDTR in proximaltubules, diphtheria toxin-induced AKI was characterized bymarked renal proximal tubular cell apoptosis with tubule dila-tion, loss of brush border, sloughing of individual epithelialcells and distal cast formation. The rate of increase in bloodurea nitrogen (BUN) and creatinine was slower than that ofischemia-reperfusion: it was observed at day 2 and peaked at5 -- 6 days. Apoptosis and regeneration were observed within2 days. Apoptosis had decreased by 5 days and evidence ofregeneration peaked at 8 days and decreased by 12 days.Complete recovery of proximal tubule function took longerand albuminuria persisted for up to 4 weeks [48].The S3 segment of the proximal tubule was selectively

targeted in transgenic mice expressing hDTR under the tran-scriptional control of the S3-specific promoter Gls5(Gsl5-hDTR) [49]. Systemic diphtheria toxin resulted in adose-dependent AKI characterized by injury mainly to theS3 segment but also to S1 and S2 segments, AQP7 expressionloss and distal cast formation.

4.2.2.2 Simultaneous targeted injury of different tubular

segmentsAnother model to selectively target tubular cells wasobtained by Cre-LoxP recombinant technology [50]. Cre-LoxP bigenic mice (Six2-GFPCre+-LoxP,iDTR+/DTRrec)expressing the diphtheria toxin receptor (iDTR+) inmetanephric-derived cells (Six2+), including proximal anddistal tubules, podocytes and the loop of Henle, suffer AKI

with diffuse tubular injury but recover completely followinga single challenge with diphtheria toxin. However, repeateddoses resulted in maladaptive repair with interstitial capillaryloss, fibrosis, and glomerulosclerosis, showing that selectiveepithelial injury can drive CKD. This may be especially rel-evant to the clinical situation in which repeated tubularinsults may occur in the course of hospitalization. Condi-tional gene expression systems, such as the tetracycline-dependent (Tet) on system, have also been employed to tar-get tubular epithelia. The Pax8/rtTA transgenic mouse is amodel of conditional gene expression created to study renaldamage that allow targeting genes to all proximal and distaltubules and the entire collecting duct system of both em-bryonic and adult kidneys [51]. On the basis of the exclusiverenal Pax8 promoter stimulation, tubular epithelial cellsexpress the reverse tetracycline-dependent transactivator(rtTA). Mice of this genetic background were crossed withTetO-X transgenic to generate double transgenicPax8-rtTA/TetO-X, where X represents different target pro-teins of interest, the expression of which is activated undertetracycline dosage. This mouse was used to model renalcancer, cystic and fibrotic lesions and it is potentially usefulin testing molecules mediators that could play a role in AKI.

4.2.2.3 Targeted injury of the thick ascending limb of the

loop of HenleThe contribution of the thick ascending limb to AKI wasinvestigated in transgenic mice expressing a herpes simplexvirus I thymidine kinase gene (HSV1-tk) commanded bythe Tamm--Horsfall promoter THP (THP-HSV1-tk), a spe-cific protein synthesized in this nephron portion [52]. Micecarrying the THP-HSV1-tk transgene are susceptible to gan-ciclovir apoptotic action whereby they showed in thickascending limbs, apoptosis, flattening of cells, dilation oftubular lumen along with cast formation, decreased renalfunction established by serum urea and creatinine rise andloss of ability to concentrate urine, all of which defined theAKI occurrence. Conditional tubular expression or deletionmay be obtained. As an example, mice with specific tubulardeletion of the focal adhesion kinase were designed to studycontributions of this signaling pathway in mediating tubulardamage in AKI [53].

4.2.3 Lineage tracingGenetically modified mice can also be used to trace cell line-age in the course of AKI or to image tubular cell stress.Genetic labeling and fate mapping of renal epithelial cells inAKI models evolving to chronicity and kidney fibrosis, suchas ischemia-reperfusion or unilateral ureteral obstruction(UUO), by Cre/Lox techniques showed no direct transitionof epithelial cells to fibroblasts [54].

4.2.4 Imaging of injured tubular segmentsNgal is a biomarker of AKI that may be assessed in humanurine. Ngal reporter mice allowed illumination of injured

A. Sanz et al.

8 Expert Opin. Drug Discov. [Early Online]

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 9: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

tubules in vivo in real time. Specific cells of the distal nephronwere the source of Ngal. This system allowed characterizingthe action of injurious and therapeutic drugs [55].

4.3 Use of AKI models to better understand the AKI

to CKD transitionThere is a growing concern about long-term consequences ofAKI that clinically may result in CKD evolving to end-stage renal disease (ESRD) [56-58]. Animal models haveallowed the exploration of the impact of AKI on the develop-ment and progression of CKD [59-62]. Modifications of thetraditional ischemia-reperfusion, folic acid or aristolochicacid-induced AKI models are commonly used to explore thetransition to chronicity [22,63-65]. UUO induces AKI, althoughit is not classically considered as a model of AKI, because earlycompensatory mechanisms of the non-ligated kidney maskloss of function of the obstructed kidney. This precludesassessment of the impact of therapy in renal function. How-ever, the extended early damage produced in the course ofan UUO is found especially useful in investigating the transi-tion to chronicity [21,66]. Fibrosis can also occur when renalinjury is mild and renal function loss is not evident [65].Depending on the dose of a unique insult or on the repetitivenature of the insult, ischemia-reperfusion, folic acid or aristo-lochic acid-induced AKI may lead to either recovery of kidneyfunction preceded by a rapid regenerative stage after the acuteinsult or chronic inflammation and fibrosis. Unraveling thecellular and molecular mechanisms may be useful in prevent-ing both the AKI to CKD transition and progression of CKDitself. A single dose of folic acid in mice resulted in fibrosisthat increased from 7 to 147 days [67]. Repeated diphtheriatoxin administration to genetically modified mice expressingthe diphtheria toxin receptor in proximal tubules also resultedin tubulointerstitial scarring associated with glomerulosclero-sis and areas of inflammatory infiltrates, 5 weeks afterAKI [50].

4.4 Novel rodent models that reproduce special

high-risk situations for AKI initiation and

progressionAKI incidence, severity and progression to CKD are more fre-quent in patients with diabetes and in the elderly [68-70]. Thus,several models of AKI have been developed in mice or ratsthat are either diabetic or aged in order to better understandthe molecular mechanisms of this predisposition and whetherthese patients require specific therapeutic approaches. Sus-ceptibility to AKI was observed in streptozotocin-induceddiabetic rats and mice [71] and septic aged mice [72]. High glu-cose and glucose degradation products promote tubular cellapoptosis and inflammation and thus may contribute to thehigher incidence of AKI in diabetics [73-76]. In mice, the con-tribution of aging-related cumulative telomere shortening tosusceptibility to apoptosis and reduced proliferative kidneyresponses to extrinsic stress have also been observed in

telomerase-deficient mice subjected to ischemic AKI. Thesemice showed increased histopathological damage and reducedproliferation of tubular, glomerular and interstitial cells com-pared to mice with mild or no telomere deficiency [77]. A roleof the antiproliferative Zag protein in preventing regenerationafter ischemia/reperfusion-induced AKI in aged mice was alsoestablished [78].

4.5 Non-rodent AKI modelsAlthough rodent models have constituted a breakthrough inunderstanding the basic processes characterizing importantgenetic as well as acquired kidney diseases, the results obtainedwith them are not exempt from misinterpretation. This is espe-cially true for conclusions from preclinical therapies that ulti-mately failed in clinical trials. Non-rodent animal models ofAKI are available and involve both larger animals that presentsome technical and physiological advantages and smaller ani-mals that better suit for conducting high-throughput chemicalor mutagenesis screens as recently reviewed [79]. Large animals,such as pigs, have similarities in size, physiology and anatomywith humans and may offer advantages to develop and validatetherapeutic approaches and transfer them into clinical trials.The growing knowledge about pig genetics has also allowedthe development of genetically modified pigs with potentialuse in research and clinical application [80]. However, applica-tion of genetically modified pig models to study AKI is stillnascent. As an example, the design of strategies blocking renalgenes identified as responsible for rejecting or inducing theexpression of other protective may potentially be useful inavoiding graft AKI in renal xenotransplants [81]. Transgenicpigs expressing the human protective HO-1 protein that mayhelp withstand ischemic insults were recently reported [82].Pigs are well suited to explore ischemia-reperfusion-inducedAKI in the context of major surgery such as renal transplanta-tion and cardiovascular surgery involving cardiopulmonarybypass [83]. Swine have also been used as a surrogate model toreproduce ischemia through maneuvers different from cardio-pulmonary bypass [84,85]. Swine is also suitable to reproduceseptic AKI and is proposed as having advantages over otheranimal models to test preclinical therapies. Production of mul-tiorgan failure with kidney compromise is not always observedin septic AKI models. A pig model combining surgical proce-dures to produce sepsis and gut ischemia/reperfusion was suc-cessful in achieving both systemic inflammation anddysfunction of the major organ systems that is typically seenin human sepsis [86]. Another model of septic AKI was alsosuitable to reproduce and study the impact of the decreasedrenal blood flow and inflammation, which have been sug-gested as mechanisms of kidney dysfunction [87]. Pig AKImodels may be used to search for molecular mediators of dis-tinctive pathological processes of AKI, such as tubular apopto-sis, as well as to study early damage biomarkers and theregeneration process accompanying recovery from AKI [88-90].

Smaller animals have also been used to model kidney dis-ease, including the worm (Caenorhabditis elegans), the fruit

Progress in the development of animal models of AKI and its impact on drug discovery

Expert Opin. Drug Discov. [Early Online] 9

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 10: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

fly (Drosophila melanogaster), the zebrafish (Danio rerio) andthe little skate (Leucoraja erinacea) [91]. In particular, malpigh-ian tubules of Drosophila are used to learn about adult kidneystem cells [92]. Gentamicin and cisplatin nephrotoxicity andAKI have been reproduced in larval zebrafish and have beenshown to respond to therapeutic intervention with a specificinhibitor of the apoptosis-related protease Omi/HtrA2 [91].Contrary to mammals, zebrafish add nephrons throughouttheir lifespan and regenerate nephrons de novo after injury,providing a model for understanding how mammalian renalregeneration may be therapeutically activated. Transplanta-tion of single aggregates comprising 10 -- 30 cells is sufficientto engraft adults and generate multiple nephrons, demonstrat-ing the presence of self-renewing nephron stem/progenitorcells in zebrafish kidneys [93].

5. New potential therapeutic approaches toAKI characterized in experimental models

The review of all potential therapeutic targets identified inrecent years in animal models of AKI is beyond the scope ofthis review. In our opinion the greatest advances are relatedto the modes of tubular cell death, the role of TWEAK inkidney inflammation and tubular cell death, the transitionto fibrosis and the recruitment and role of macrophages andmarrow cells.Tubular cell death is a key feature of AKI. Both apoptosis

and necrosis contribute to tubular cell injury and the relativecontribution may depend on the severity of injury and thespecific trigger [7,9]. The role of apoptosis has been extensivelystudied but has not yet resulted in any medication in clinicaluse. By contrast, necrosis was neglected because it was thoughtto be a passive process not susceptible to therapeutic manipu-lation. This view has changed in recent years and there is hopethat a particular form of regulated necrosis, necroptosis,

which is controlled by the kinases RIPK1 and RIPK3, mayprovide novel therapeutic tools [94]. Necroptosis may beinhibited by necrostatin-1, a highly specific inhibitor ofRIPK1. Necrostatin-1 reduced kidney damage and renal fail-ure, even when administered after reperfusion, resulting in asignificant survival benefit in a model of lethal renal ische-mia/reperfusion injury in which the pan-caspase inhibitorzVAD, an inhibitor of apoptosis, was not protective [95].

Two novel therapeutic targets encompass the cell death--inflammation interface. A polymeric nanomedicine, whichwas previously shown to inhibit cell death induced by theproapoptotic molecule Apaf-1, was recently shown to preventtubular cell death and inflammatory responses both in cultureand during folic acid-induced AKI in mice [96,97]. TWEAK isa TNF superfamily cytokine that promotes tubular cellinflammatory responses and tubular cell death through activa-tion of the Fn14 receptor [8,98]. TWEAK or Fn14 targeting byantibodies or in KO mice prevents folic acid and ischemia-reperfusion-induced AKI, tubular cell apoptosis andinflammation [8,41,42,99-101]. Interestingly TWEAK targetingpreserved the kidney expression of Klotho during AKI [102].Klotho behaves both as an FGF23 receptor and a soluble hor-mone with anti-aging, anti-inflammatory and anti-fibroticproperties [103,104].

Fibrosis is a general feature of CKD and a determinant ofprogression to ESRD [105]. The repair process after AKI canbe incomplete, with persisting tubulointerstitial inflammationand fibrosis characterized by proliferation of fibroblasts andexcessive deposition of extracellular matrix [106]. The molecu-lar mechanisms that mediate renal fibrosis after AKI are notwell understood. AKI animal models have been used to dissectthe molecular mechanisms and find new therapeutic tar-get [107]. Recent studies have shown that fibrosis is promotedby tubular cell G2/M cell cycle arrest during AKI or even dur-ing milder renal injury not leading to serum creatinine

Table 3. Molecular and biological pathways targeted in recent or ongoing AKI clinical trials.

Candidate target Model Species Targeting procedure Phenotypic responses Renal

function

Clinical

trial

Refs.

Multipotent MSCs IRI Rat AC607 # Tubular apoptosisand inflammation

Improved Ongoing [124]

Oxidative stress IRI Rat Administration of N-AC(inhibition of endothelialnitric oxide synthase)

# Infiltrating leukocytes Non-significanttrend towardsimprovement

Completedand ongoing

[134]

Apoptosis IRI Rat Administration of EPO # Tubular damage,apoptosis and NF-kBimmunostaining

Improved Completedand ongoing

[135]

Vessel vasoconstriction IRI Rat Administration of ANP # Acute tubular necrosis Improved Completed [136]

Dopamine (D-1)receptor

IRI Rat Administration offenoldopam, D-1 agonist

# Activation of NF-kB Not assessed Completedand ongoing

[137]

Dopamine (D-1)receptor

IRI Pig Administration offenoldopam, D-1 agonist

# Acute tubular necrosis Improved Completedand ongoing

[138]

p53 IRI andcisplatin

Rat I5NP: p53 siRNA # Tubular apoptosis Improved Ongoing [139]

IRI: Ischemia-reperfusion injury.

A. Sanz et al.

10 Expert Opin. Drug Discov. [Early Online]

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 11: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

elevations [22]. G2/M-arrested proximal tubular cells activateJNK signaling, leading to the release of profibrotic cytokines,such as TGF-b1 and CTGF, which in turn stimulated prolif-eration and collagen synthesis by fibroblasts. These studiesidentified two potential druggable targets: JNK inhibition orbypassing the G2/M arrest by administration of a p53inhibitor prevented fibrosis. The process was observed afterkidney injury induced by severe or unilateral ischemia/re-per-fusion injury, aristolochic acid nephropathy and UUO [22].Indeed, p53 targeting is already undergoing clinical trials inAKI (see below) and the oral JNK inhibitor is undergoingclinical trials for inflammatory endometriosis (clinicaltrials.gov/ct2/show/NCT01630252). Demonstration of safetymay facilitate its assessment in AKI o renal fibrosis.

UUO and ischemia-reperfusion AKI illustrated the poten-tial therapeutic interest of THR-123 to prevent progressionto fibrosis. THR-123 is a peptide agonist of the activin-like kinase 3 (Alk3) receptor. BMP7 antagonizes the TGFb/Smad pathway by binding to Alk3 [66,108]. THR-123 repro-duced BMP7 actions and decreased tubular damage andfibrosis, but did not prevent the increase in BUN followingischemia-reperfusion [66].

HIPK2 is a protein kinase identified as a kidney fibrosispromoter by a systems biology approach in the fibrotic kid-neys of Tg26 mice transgenic for HIV [109]. The role ofHIPK2 in kidney fibrosis was validated in murine UUOand folic acid-induced renal fibrosis. Absence of HIPK2 pre-vented both fibrosis and loss of renal function in these mod-els [109]. As other kinases, HIPK2 is a therapeutic targetpotentially druggable by oral small molecules.

The contribution of methylation epigenetic modificationsto kidney fibrosis was established in kidney injury inducedby a single dose of folic acid [67]. Fibrosis was reduced in het-erozygous mice by the expression of the methyltransferaseDnmt, suggesting that this enzyme is a potential therapeutictarget. Cell culture indicated that methylation of the Rasregulatory molecule Rasal1 played a key role.

Several studies have addressed the role of CXCR4 andCXCR7, the two receptors for the chemokine stromal-derivedfactor-1 (SDF-1), in AKI. In SCID mice, rhabdomyolysis-induced AKI following glycerol injection CXCR4 andCXCR7 played an essential role in the therapeutic homingof human renal progenitor cells, tissue regeneration and renalfunction improvement [110]. This has important implicationsfor the development of stem cell-based therapies. A note ofcaution is needed since in a model of Stx2-induced AKI inCAST/Ei mice, inhibition of the CXCR4--SDF-1 interactiondecreased endothelial activation and organ injury andimproved animal survival [26]. Thus, CXCR4/SDF-1 targetingprotects from experimental HUS-induced AKI but mayimpair homing of renal progenitor cells.

Marrow cells also contribute to AKI recovery. A role of res-ident renal macrophages and dendritic cells in AKI recoverywas observed after ischemia/reperfusion injury or followingdiphtheria toxin selective injury to proximal tubules [48].

Macrophage/dendritic cell depletion during the recoveryphase increased functional and histological injury and delayedregeneration. Genetic or pharmacological inhibition of mac-rophage colony-stimulating factor (CSF-1) signaling blockedmacrophage/dendritic cell proliferation, decreased M2 polari-zation and inhibited recovery. These findings demonstratedthat CSF-1-mediated expansion and polarization of residentrenal macrophages/dendritic cells is an important mechanismmediating renal tubule epithelial regeneration after AKI [48].

Multiple myeloma is a plasma cell dyscrasia in which over-production of light chains may result in AKI and CKD causeda light chain precipitation in association with Tamm--Horsfallprotein into the tubular lumen (cast nephropathy). The mainprognostic factor for cast nephropathy is the response of thetumor to chemotherapy. An unmet medical need is the treat-ment of non-responsive patients. Cast nephropathy and AKIwere reproduced in rats 72 h after a single intraperitonealinjection of monoclonal light chains (k3 or l2) obtainedfrom myeloma patients with cast nephropathy [111]. Thismodel was used to illustrate protection from casts and AKIprovided by a cyclized competitor peptide that inhibited thebinding of light chains to Tamm--Horsfall protein, thusproviding the first therapy for cast nephropathy that isindependent from the tumor response to treatment [112].

6. Clinical therapies for AKI treatment

Current clinical strategies for AKI prevention are limited torestriction of nephrotoxic agents and avoiding hypoperfusionand volume depletion. However, there are no established ther-apies for AKI beyond symptomatic management. The currentstatus of clinical trials in AKI has been recently reviewed [113].As of November 2011, there were 126 trials registered onclinicalTrials.org studying AKI: 75 addressed prevention ofAKI and 51 addressed management of established AKI. Themajority of the prevention trials are conducted for cardiacsurgery-associated AKI and contrast-induced AKI [113-115].N-acetylcysteine (NAC), erythropoietin (EPO), natriureticpeptides, fenoldopam, I5NP and AC607 are some of theagents undergoing clinical testing in AKI based on resultsfrom experimental animals (Table 3). Neutralizing anti-TWEAK antibodies are not undergoing clinical trials inAKI, but there is a first human clinical trial addressing itsnephroprotective action in lupus nephritis, and if this trialsucceeds, we might expect trials in AKI to follow.

NAC has been studied for AKI prevention, since 2000.However, results have been mixed [116]. The PRESERVEstudy (Prevention of Serious Adverse Events FollowingAngiography) is enrolling 8,680 patients to settle theissue [117].

EPO is an erythropoietic hormone produced by the kidney.Accumulating evidence suggests that EPO has additionalorgan protective effects [118]. A randomized, pilot study ofpreoperative EPO in 71 patients who underwent elective cor-onary artery bypass graft surgery reported renoprotective

Progress in the development of animal models of AKI and its impact on drug discovery

Expert Opin. Drug Discov. [Early Online] 11

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 12: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

effects [119]. In a different setting, randomization of 162 ICUpatients at risk of AKI based on urinary biomarkers (urinaryGGT and alkaline phosphatase) to placebo or two doses ofEPO did not result in differences in the incidence ofAKI [118]. There are currently three randomized controlledtrials using preoperative EPO.Although there are currently no clinical trials testing car-

peritide (atrial natriuretic peptide) and nesiritide (brain natri-uretic peptide), they are worth mentioning as examples ofpromising preclinical data not mirrored in the clinical setting.A recent meta-analysis comprising data from 15 randomized,controlled trials in cardiovascular surgery patients reported anincreased urine output and a reduction in serum creatininelevels compared with controls. However, the effect isdescribed in the immediate perioperative period and noconclusions can be drawn on outcomes [120].Fenoldopam is a dopaminergic DA1 agonist that selectively

increases renal medullary blood flow and natriuresis and hasbeen tested in clinical trials as prophylaxis for septic, postop-erative and contrast-induced AKI with no clear benefits [121].Fenoldopam is currently being tested as a prophylactic agentfor AKI in cardiopulmonary bypass surgery.I5NP is a synthetic RNAi that temporarily inhibits the

expression of p53. I5NP is being developed to protect cellsfrom acute ischemia/reperfusion injuries and AKI followingmajor cardiac surgery and renal transplantation [122]. Thep53 is a proapoptotic protein that additionally promotes G2/M cell cycle arrest and kidney fibrosis in murine AKI mod-els [7,22]. I5NP was tested in a Phase I clinical trial in 16 patientsat risk of AKI who had undergone cardiovascular surgery. In aPhase II trial, I5NP is being tested in patients receivingdeceased donor kidney transplantation with > 24 h of coldischemia time, to evaluate efficacy in preventing delayed graftfunction. However, a dose escalation and safety study ofI5NP to prevent AKI in high-risk major cardiovascular surgerypatients was terminated [123].AC607 is composed of human bone marrow-derived

mesenchymal stem cells (MSCs). In preclinical research, itdecreased inflammation and cell death in anesthetized ratswith ischemia-reperfusion-induced AKI [124]. An ongoingclinical trial is testing effects of AC607 in AKI patients within48 h of cardiac surgery. Once enrolled, subjects will receive asingle administration of AC607 or placebo. Kidney recoverywill be evaluated over the subsequent 30 days and death orthe need for dialysis will be evaluated within 90 days of dos-ing. After 90 days (evaluation period), subjects will enter a3-year extension phase of the study to monitor safety andlong-term outcomes (follow-up period) [125].

7. Conclusions

In conclusion, while the yield to date of animal models hasbeen poor as assessed by the number of drugs approved forthe treatment of human AKI, there is a new crop of optimizedmodels and it is conceivable that further refinement of animal

models will eventually result in effective therapies for clinicalAKI. It should be kept in mind, however, that these modelsshould be used to test the therapeutic rather than prophylacticuse of agents, given that in the clinic, many patients presentwith already established AKI. In addition, advances in bio-marker science should be incorporated to adequately stageAKI and individualize therapy according to AKI stage.

8. Expert opinion

The number of experimental AKI models has increased inrecent years trying to address concerns resulting from thepoor performance of prior therapeutic targets identified in con-ventional AKI models when translated to clinical practice andalso trying to keep pace with newly described entities. In addi-tion, the number of species used for AKI models has expandedto include non-rodents. Non-rodents allow the evaluation ofspecific aspects of AKI in bigger animals, such as complex sur-gical interventions and xenograft protection from AKI in pigsand large-scale genetic screenings in simpler organisms suchas drosophila and zebrafish. New rodent models have repro-duced recently described clinical entities, such as aristolochicand warfarin nephropathies, have provided better models forold entities such as thrombotic microangiopathy-induced AKIor cast nephropathy, have optimized models to explore factorsthat predispose AKI in clinical practice such as prior CKD,old age or diabetes and have modified the model to answer spe-cific questions such as the factors driving the AKI to CKD tran-sition. Furthermore, genetically modified animals have beendesigned to explore functional contributors to AKI, to help tar-get specific cell types and to track the contribution of specificcell types. A combination of traditional and new models hasallowed the identification of novel druggable targets that mod-ulate the development of AKI or try to correct the failure torecover from injury. These new targets include, but are not lim-ited to, modulators of cell death, such as RIP1/RIP3 regulatingnecroptosis, inflammatory cytokines, such as TWEAK, media-tors of the AKI to CKD transition, such as JNK, Alk3,HIPK2 and Dnmt and regulators of bone marrow cells andstem cell biology, such as CXCR4/CXCR7/SDF-1 andCSF-1. Agents targeting some of these molecules are alreadyundergoing clinical trials. Thus, the deep understanding ofthe role of TWEAK/Fn14 in kidney disease justifies the factthat the first clinical trial addressing the efficacy of neutralizinganti-TWEAK antibodies in humans studies nephroprotectionin lupus nephritis. An eventual success of this trial may openthe doors for studies in AKI. JNK, Alk3, HIPK2 and Dnmtrepresent receptors and enzymes which can be inhibited bysmall molecules, some of which, such as JNK inhibitors, areundergoing clinical trials for inflammatory diseases [126]. Atleast 17 trials are studying or have studied CXCR4 inhibitorsor antagonists for diverse clinical indications [127]. Furthermore,several therapies identified in AKI animal models are undergo-ing clinical trials in human AKI, including p53 RNAi andbone-marrow derived MSCs.

A. Sanz et al.

12 Expert Opin. Drug Discov. [Early Online]

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 13: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

Acknowledgements

The authors have received support from the Instituto de SaludCarlos III through the grants FIS PS09/00447, ISCIII-RETIC, REDinREN/RD06/0016. Furthermore, they havereceived support via the European Regional DevelopmentFund (FEDER), RD12/0021 and the Comunidad deMadrid/CIFRA S2010/BMD-2378. The authors have alsoreceived support via the Instituto de Salud Carlos III in theform of: MD Sanchez-Nino has received a FIS-Sara Borrell

post-doctoral fellowship; AM Ramos and A Sanz have receiveda post-doctoral Miguel Servet fellowship, while A Ortiz hasreceived salary support through the Programa IntensificacionActividad Investigadora (ISCIII/Agencia Laın-Entralgo/CM).Finally C Martin-Cleary is the recipient of a ‘Rio Hortega’fellowship grant from the Instituto de Salud Carlos III.

Declaration of Interest

The authors have no conflict of Interest

BibliographyPapers of special note have been highlighted as

either of interest (�) or of considerable interest(��) to readers.

1. Bellomo R, Kellum JA, Ronco C. Acute

kidney injury. Lancet

2012;380(9843):756-66

2. Van Berendoncks AM, Elseviers MM,

Lins RL, Group SS. Outcome of acute

kidney injury with different treatment

options: long-term follow-up. Clin J Am

Soc Nephrol 2010;5(10):1755-62

3. Hoste EA, Clermont G, Kersten A, et al.

RIFLE criteria for acute kidney injury are

associated with hospital mortality in

critically ill patients: a cohort analysis.

Crit Care 2006;10(3):R73

4. Xue JL, Eggers PW, Agodoa LY, et al.

Longitudinal study of racial and ethnic

differences in developing end-stage renal

disease among aged medicare

beneficiaries. J Am Soc Nephrol

2007;18(4):1299-306

5. Hsu CY, McCulloch CE, Fan D, et al.

Community-based incidence of acute

renal failure. Kidney Int

2007;72(2):208-12

6. Chawla LS, Kimmel PL. Acute kidney

injury and chronic kidney disease:

an integrated clinical syndrome.

Kidney Int 2012;82(5):516-24

7. Sanz AB, Santamarıa B, Ruiz-Ortega M,

et al. Mechanisms of renal apoptosis in

health and disease. J Am Soc Nephrol

2008;19(9):1634-42.. An updated overview of the regulation

and contribution of apoptosis to AKI.

8. Sanz AB, Justo P, Sanchez-Nino MD,

et al. The cytokine TWEAK modulates

renal tubulointerstitial inflammation.

J Am Soc Nephrol 2008;19(4):695-703.. First description of the theraupeutic

potential of TWEAK targeting in

kidney injury. Anti-TWEAK antibodies

are undergoing clinical trials in

inflammatory kidney injury.

9. Ortiz A, Justo P, Sanz A, et al. Targeting

apoptosis in acute tubular injury.

Biochem Pharmacol 2003;66(8):1589-94

10. Endre ZH, Pickering JW, Walker RJ,

et al. Improved performance of urinary

biomarkers of acute kidney injury in the

critically ill by stratification for injury

duration and baseline renal function.

Kidney Int 2011;79(10):1119-30

11. Heyman SN, Lieberthal W, Rogiers P,

et al. Animal models of acute tubular

necrosis. Curr Opin Crit Care

2002;8(6):526-34.. This review highlights general principles

in planning, conducting and critically

evaluating studies using animal models.

It focuses on the principles, advantages

and disadvantages of selected AKI

animal models and gathers the expert

opinion of four relevant researchers in

the renal area.

12. Singh AP, Junemann A, Muthuraman A,

et al. Animal models of acute renal

failure. Pharmacol Rep 2012;64(1):31-44. This review provides a complete list of

AKI models and specifies relevant

technical aspects for each one of them.

13. Waterston RH, Lindblad-Toh K,

Birney E, et al. Initial sequencing and

comparative analysis of the mouse

genome. Nature 2002;420(6915):520-62

14. Gibbs RA, Weinstock GM, Metzker ML,

et al. Genome sequence of the Brown

Norway rat yields insights into

mammalian evolution. Nature

2004;428(6982):493-521

15. Dwinell MR, Lazar J, Geurts AM. The

emerging role for rat models in gene

discovery. Mamm Genome

2011;22(7-8):466-75

16. Basile DP, Dwinell MR, Wang SJ, et al.

Chromosome substitution modulates

resistance to ischemia reperfusion injury

in Brown Norway rats. Kidney Int

2013;83(2):242-50

17. Mengs U. Acute toxicity of aristolochic

acid in rodents. Arch Toxicol

1987;59(5):328-31

18. Cosyns JP, Jadoul M, Squifflet JP, et al.

Chinese herbs nephropathy: a clue to

Balkan endemic nephropathy? Kidney Int

1994;45(6):1680-8

19. Cosyns JP, Dehoux JP, Guiot Y, et al.

Chronic aristolochic acid toxicity in

rabbits: a model of Chinese herbs

nephropathy? Kidney Int

2001;59(6):2164-73

20. Debelle FD, Nortier JL, De Prez EG,

et al. Aristolochic acids induce chronic

renal failure with interstitial fibrosis in

salt-depleted rats. J Am Soc Nephrol

2002;13(2):431-6

21. De Broe ME. Chinese herbs nephropathy

and Balkan endemic nephropathy:

toward a single entity, aristolochic acid

nephropathy. Kidney Int

2012;81(6):513-15. Updated review of the role of

aristolochic acid in kidney injury.

22. Yang L, Besschetnova TY, Brooks CR,

et al. Epithelial cell cycle arrest in G2/M

mediates kidney fibrosis after injury.

Nat Med 2010;16(5):535-43

23. Zager RA, Johnson AC, Naito M, et al.

Maleate nephrotoxicity: mechanisms of

injury and correlates with ischemic/

hypoxic tubular cell death. Am J Physiol

Renal Physiol 2008;294(1):F187-97

24. Schmidtko J, Peine S, El-Housseini Y,

et al. Treatment of atypical hemolytic

uremic syndrome and thrombotic

microangiopathies: a focus on

eculizumab. Am J Kidney Dis

2013;61(2):289-99

25. Menne J, Nitschke M, Stingele R, et al.

Validation of treatment strategies for

Progress in the development of animal models of AKI and its impact on drug discovery

Expert Opin. Drug Discov. [Early Online] 13

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 14: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

enterohaemorrhagic Escherichia coli

O104:H4 induced haemolytic uraemic

syndrome: case-control study. BMJ

2012;345:e4565

26. Petruzziello-Pellegrini TN, Yuen DA,

Page AV, et al. The CXCR4/CXCR7/

SDF-1 pathway contributes to the

pathogenesis of Shiga toxin-associated

hemolytic uremic syndrome in humans

and mice. J Clin Invest

2012;122(2):759-76

27. Mallick EM, McBee ME, Vanguri VK,

et al. A novel murine infection model for

Shiga toxin-producing Escherichia coli.

J Clin Invest 2012;122(11):4012-24. Description of a novel model of AKI

due to infectious thrombotic

microangipathy which shares features

with the human condition.

28. Hohenstein B, Braun A, Amann KU,

et al. A murine model of site-specific

renal microvascular endothelial injury

and thrombotic microangiopathy.

Nephrol Dial Transplant

2008;23(4):1144-56

29. Psotka MA, Obata F, Kolling GL, et al.

Shiga toxin 2 targets the murine renal

collecting duct epithelium. Infect Immun

2009;77(3):959-69

30. Ware K, Brodsky P, Satoskar AA, et al.

Warfarin-related nephropathy modeled

by nephron reduction and excessive

anticoagulation. J Am Soc Nephrol

2011;22(10):1856-62. Description of an animal model for a

newly recognized form of AKI in

excessively anticoagulated patients.

31. Rizk DV, Warnock DG.

Warfarin-related nephropathy: another

newly recognized complication of an old

drug. Kidney Int 2011;80(2):131-3

32. Gutierrez E, Egido J, Rubio-Navarro A,

et al. Oxidative stress, macrophage

infiltration and CD163 expression are

determinants of long-term renal outcome

in macrohematuria-induced acute kidney

injury of IgA nephropathy.

Nephron Clin Pract

2012;121(1-2):c42-53

33. Moreno JA, Martın-Cleary C,

Gutierrez E, et al. AKI associated with

macroscopic glomerular hematuria:

clinical and pathophysiologic

consequences. Clin J Am Soc Nephrol

2012;7(1):175-84. A condensed review of

hemoglobin-induced AKI in the

context of glomerular injury.

34. Moreno JA, Martın-Cleary C,

Gutierrez E, et al. Haematuria: the

forgotten CKD factor?

Nephrol Dial Transplant

2012;27(1):28-34

35. Martın Cleary C, Moreno JA,

Fernandez B, et al. Glomerular

haematuria, renal interstitial haemorrhage

and acute kidney injury.

Nephrol Dial Transplant

2010;25(12):4103-6

36. Torres AM, Dnyanmote AV, Bush KT,

et al. Deletion of multispecific organic

anion transporter Oat1/Slc22a6 protects

against mercury-induced kidney injury.

J Biol Chem 2011;286(30):26391-5

37. Nath KA, Croatt AJ, Warner GM, et al.

Genetic deficiency of Smad3 protects

against murine ischemic acute kidney

injury. Am J Physiol Renal Physiol

2011;301(2):F436-42

38. Bengatta S, Arnould C, Letavernier E,

et al. MMP9 and SCF protect from

apoptosis in acute kidney injury. J Am

Soc Nephrol 2009;20(4):787-97

39. Wei Q, Bhatt K, He HZ, et al. Targeted

deletion of Dicer from proximal tubules

protects against renal

ischemia-reperfusion injury. J Am

Soc Nephrol 2010;21(5):756-61

40. Furuichi K, Gao JL, Horuk R, et al.

Chemokine receptor CCR1 regulates

inflammatory cell infiltration after renal

ischemia-reperfusion injury. J Immunol

2008;181(12):8670-6

41. Sanz AB, Sanchez-Nino MD,

Izquierdo MC, et al. Tweak induces

proliferation in renal tubular epithelium:

a role in uninephrectomy induced renal

hyperplasia. J Cell Mol Med

2009;13(9B):3329-42

42. Hotta K, Sho M, Yamato I, et al. Direct

targeting of fibroblast growth

factor-inducible 14 protein protects

against renal ischemia reperfusion injury.

Kidney Int 2011;79(2):179-88

43. Li HF, Cheng CF, Liao WJ, et al.

ATF3-mediated epigenetic regulation

protects against acute kidney injury.

J Am Soc Nephrol 2010;21(6):1003-13

44. Kim J, Zarjou A, Traylor AM, et al. In

vivo regulation of the heme

oxygenase-1 gene in humanized

transgenic mice. Kidney Int

2012;82(3):278-91

45. de Jorge EG, Macor P,

Paixao-Cavalcante D, et al. The

development of atypical hemolytic

uremic syndrome depends on

complement C5. J Am Soc Nephrol

2011;22(1):137-45

46. Ortiz A, Justo P, Sanz A, et al. Tubular

cell apoptosis and cidofovir-induced

acute renal failure. Antivir Ther

2005;10(1):185-90

47. Fernandez-Fernandez B,

Montoya-Ferrer A, Sanz AB, et al.

Tenofovir nephrotoxicity: 2011 update.

AIDS Res Treat 2011;2011:354908

48. Zhang MZ, Yao B, Yang S, et al.

CSF-1 signaling mediates recovery from

acute kidney injury. J Clin Invest

2012;122(12):4519-32

49. Sekine M, Monkawa T, Morizane R,

et al. Selective depletion of mouse kidney

proximal straight tubule cells causes acute

kidney injury. Transgenic Res

2012;21(1):51-62

50. Grgic I, Campanholle G, Bijol V, et al.

Targeted proximal tubule injury triggers

interstitial fibrosis and glomerulosclerosis.

Kidney Int 2012;82(2):172-83.. Illustrates how the advanced genetic

engineering can be used to design a

highly selective mouse model of

tubular injury to define the acute and

chronic repercussion of isolated

epithelial injury.

51. Traykova-Brauch M, Sch€onig K,

Greiner O, et al. An efficient and

versatile system for acute and chronic

modulation of renal tubular function in

transgenic mice. Nat Med

2008;14(9):979-84. Description of transgenic mice that

allow expression of specific genes in all

proximal and distal tubules and the

entire collecting duct system of both

embryonic and adult kidneys.

52. Srichai MB, Hao C, Davis L, et al.

Apoptosis of the thick ascending limb

results in acute kidney injury. J Am

Soc Nephrol 2008;19(8):1538-46

53. Qin Y, Alderliesten MC, Stokman G,

et al. Focal adhesion kinase signaling

mediates acute renal injury induced by

ischemia/reperfusion. Am J Pathol

2011;179(6):2766-78

54. Humphreys BD, Lin SL, Kobayashi A,

et al. Fate tracing reveals the pericyte and

not epithelial origin of myofibroblasts in

kidney fibrosis. Am J Pathol

2010;176(1):85-97.. Description of genetically modified

mice as tools for lineage tracing in the

A. Sanz et al.

14 Expert Opin. Drug Discov. [Early Online]

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 15: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

course of AKI evolving to renal

fibrosis -- in search of the origin of the

kidney fibroblast.

55. Paragas N, Qiu A, Zhang Q, et al. The

Ngal reporter mouse detects the response

of the kidney to injury in real time.

Nat Med 2011;17(2):216-22.. Characterization of genetically

modified mice that allows imaging of

tubular cell injury in real time. This

advantage more precisely allows

establishing the initial time of damage

which may result in shortening the

effective therapeutic time frame.

56. Leung KC, Tonelli M, James MT.

Chronic kidney disease following acute

kidney injury-risk and outcomes.

Nat Rev Nephrol 2013;9(2):77-85

57. Coca SG, Singanamala S, Parikh CR.

Chronic kidney disease after acute kidney

injury: a systematic review and

meta-analysis. Kidney Int 2012;81(5):442-8

58. Bydash JR, Ishani A. Acute kidney injury

and chronic kidney disease: a work in

progress. Clin J Am Soc Nephrol

2011;6(11):2555-7

59. Gueler F, Gwinner W, Schwarz A, et al.

Long-term effects of acute ischemia and

reperfusion injury. Kidney Int

2004;66(2):523-7

60. Basile DP, Leonard EC, Tonade D, et al.

Distinct effects on long-term function of

injured and contralateral kidneys

following unilateral renal

ischemia-reperfusion. Am J Physiol

Renal Physiol 2012;302(5):F625-35

61. Ortega A, Ramila D, Ardura JA, et al.

Role of parathyroid hormone-related

protein in tubulointerstitial apoptosis and

fibrosis after folic acid-induced

nephrotoxicity. J Am Soc Nephrol

2006;17(6):1594-603

62. Verdoorn KS, Lindoso RS, Lowe J, et al.

Bone marrow mononuclear cells shift

bioactive lipid pattern in injured kidney

towards tissue repair in rats with

unilateral ureteral obstruction.

Nephrol Dial Transplant

2010;25(12):3867-74

63. Fang TC, Alison MR, Cook HT, et al.

Proliferation of bone marrow-derived

cells contributes to regeneration after

folic acid-induced acute tubular injury.

J Am Soc Nephrol 2005;16(6):1723-32

64. He S, Liu N, Bayliss G, et al. EGFR

activity is required for renal tubular cell

dedifferentiation and proliferation in a

murine model of folic acid-induced acute

kidney injury. Am J Physiol

Renal Physiol 2013;304(4):F356-66

65. Bonventre JV, Yang L. Cellular

pathophysiology of ischemic acute kidney

injury. J Clin Invest 2011;121(11):4210-21.. Updated review of the cellular and

molecular mechanism underlying

ischemia-reperfusion-induced AKI.

66. Sugimoto H, LeBleu VS, Bosukonda D,

et al. Activin-like kinase 3 is important

for kidney regeneration and reversal of

fibrosis. Nat Med 2012;18(3):396-404.. Characterization of Alk3 as a

therapeutic target controlling

regeneration and fibrosis

following AKI.

67. Bechtel W, McGoohan S, Zeisberg EM,

et al. Methylation determines fibroblast

activation and fibrogenesis in the kidney.

Nat Med 2010;16(5):544-50.. Identification of epigenetic regulators

as determinants of the transition from

AKI to kidney fibrosis and as

therapeutic targets.

68. Calvin AD, Misra S, Pflueger A.

Contrast-induced acute kidney injury and

diabetic nephropathy. Nat Rev Nephrol

2010;6(11):679-88

69. Kelly KJ, Dominguez JH. Rapid

progression of diabetic nephropathy is

linked to inflammation and episodes of

acute renal failure. Am J Nephrol

2010;32(5):469-75

70. Ishani A, Xue JL, Himmelfarb J, et al.

Acute kidney injury increases risk of

ESRD among elderly. J Am Soc Nephrol

2009;20(1):223-8

71. Collino M, Benetti E, Miglio G, et al.

Peroxisome proliferator-activated receptor

b/d agonism protects the kidney against

ischemia/reperfusion injury in diabetic

rats. Free Radic Biol Med

2011;50(2):345-53

72. Maddens B, Vandendriessche B,

Demon D, et al. Severity of

sepsis-induced acute kidney injury in a

novel mouse model is age dependent.

Crit Care Med 2012;40(9):2638-46

73. Ortiz A, Ziyadeh FN, Neilson EG.

Expression of apoptosis-regulatory genes

in renal proximal tubular epithelial cells

exposed to high ambient glucose and in

diabetic kidneys. J Investig Med

1997;45(2):50-6. First description of the response of

apoptosis regulatory genes to ambient

glucose in kidney cells.

74. Justo P, Sanz AB, Egido J, et al. 3,4-

Dideoxyglucosone-3-ene induces

apoptosis in renal tubular epithelial cells.

Diabetes 2005;54(8):2424-9

75. Sanchez-Nino MD, Sanz AB, Lorz C,

et al. BASP1 promotes apoptosis in

diabetic nephropathy. J Am Soc Nephrol

2010;21(4):610-21

76. Sanchez-Nino MD, Benito-Martin A,

Ortiz A. New paradigms in cell death in

human diabetic nephropathy. Kidney Int

2010;78(8):737-44

77. Westhoff JH, Schildhorn C, Jacobi C,

et al. Telomere shortening reduces

regenerative capacity after acute kidney

injury. J Am Soc Nephrol

2010;21(2):327-36

78. Schmitt R, Marlier A, Cantley LG. Zag

expression during aging suppresses

proliferation after kidney injury. J Am

Soc Nephrol 2008;19(12):2375-83

79. Hentschel DM, Bonventre JV. Novel

non-rodent models of kidney disease.

Curr Mol Med 2005;5(5):537-46. A classical review of non-rodent

models of kidney injury. It specifically

deals with small animals to model

kidney disease and discusses specific

application and advantages over rodent

and large animal models.

80. Luo Y, Lin L, Bolund L, et al.

Genetically modified pigs for biomedical

research. J Inherit Metab Dis

2012;35(4):695-713

81. Walters EM, Wolf E, Whyte JJ, et al.

Completion of the swine genome will

simplify the production of swine as a

large animal biomedical model.

BMC Med Genomics 2012;5:55. This review deals with swine as a

biochemical model for human disease

research and provides examples of

pig-based models, including novel

application of genetically modified pigs

for the study of renal disease.

82. Yeom HJ, Koo OJ, Yang J, et al.

Generation and characterization of

human heme oxygenase-1 transgenic pigs.

PLoS One 2012;7(10):e46646

83. Patel NN, Toth T, Jones C, et al.

Prevention of post-cardiopulmonary

bypass acute kidney injury by endothelin

A receptor blockade. Crit Care Med

2011;39(4):793-802

84. Hunter JP, Hosgood SA, Patel M, et al.

Effects of hydrogen sulphide in an

experimental model of renal

Progress in the development of animal models of AKI and its impact on drug discovery

Expert Opin. Drug Discov. [Early Online] 15

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 16: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

ischaemia-reperfusion injury. Br J Surg

2012;99(12):1665-71

85. Favreau F, Rossard L, Zhang K, et al.

Expression and modulation of

translocator protein and its partners by

hypoxia reoxygenation or ischemia and

reperfusion in porcine renal models.

Am J Physiol Renal Physiol

2009;297(1):F177-90

86. Kubiak BD, Albert SP, Gatto LA, et al.

A clinically applicable porcine model of

septic and ischemia/reperfusion-induced

shock and multiple organ injury.

J Surg Res 2011;166(1):e59-69

87. Lipcsey M, Bellomo R. Septic acute

kidney injury: hemodynamic syndrome,

inflammatory disorder, or both?

Crit Care 2011;15(6):1008

88. Grossini E, Molinari C, Pollesello P,

et al. Levosimendan protection against

kidney ischemia/reperfusion injuries in

anesthetized pigs. J Pharmacol Exp Ther

2012;342(2):376-88

89. Rossard L, Favreau F, Demars J, et al.

Evaluation of early regenerative processes

in a preclinical pig model of acute

kidney injury. Curr Mol Med

2012;12(4):502-5

90. Hosgood SA, Hunter JP, Nicholson ML.

Early urinary biomarkers of warm and

cold ischemic injury in an experimental

kidney model. J Surg Res

2012;174(2):e85-90

91. Hentschel DM, Park KM, Cilenti L,

et al. Acute renal failure in zebrafish:

a novel system to study a complex

disease. Am J Physiol Renal Physiol

2005;288(5):F923-9

92. Singh SR, Hou SX. Lessons learned

about adult kidney stem cells from the

malpighian tubules of Drosophila. J Am

Soc Nephrol 2008;19(4):660-6

93. Diep CQ, Ma D, Deo RC, et al.

Identification of adult nephron

progenitors capable of kidney

regeneration in zebrafish. Nature

2011;470(7332):95-100. Characterization of zebrafish as a

model system for studying

renal regeneration.

94. Linkermann A, De Zen F, Weinberg J,

et al. Programmed necrosis in acute

kidney injury. Nephrol Dial Transplant

2012;27(9):3412-19.. Updated overview of the role and

regulation of programmed necrosis in

kidney injury.

95. Linkermann A, Brasen JH,

Himmerkus N, et al. Rip1 (receptor-

interacting protein kinase 1) mediates

necroptosis and contributes to renal

ischemia/reperfusion injury. Kidney Int

2012;81(8):751-61

96. Santamarıa B, Benito-Martin A,

Ucero AC, et al. A nanoconjugate

Apaf-1 inhibitor protects mesothelial cells

from cytokine-induced injury. PLoS One

2009;4(8):e6634

97. Ucero AC, Berzal S, Ocana-Salceda C,

et al. A polymeric nanomedicine

diminishes inflammatory events in renal

tubular cells. PLoS One

2013;8(1):e51992. Novel therapeutic agent targeting

inflammation and cell death.

98. Ortiz A, Sanz AB, Munoz Garcıa B,

et al. Considering TWEAK as a target

for therapy in renal and vascular injury.

Cytokine Growth Factor Rev

2009;20(3):251-8

99. Izquierdo MC, Sanz AB, Mezzano S,

et al. TWEAK (tumor necrosis factor-like

weak inducer of apoptosis) activates

CXCL16 expression during renal

tubulointerstitial inflammation.

Kidney Int 2012;81(11):1098-107

100. Sanz AB, Sanchez-Nino MD,

Izquierdo MC, et al. TWEAK activates

the non-canonical NFkappaB pathway in

murine renal tubular cells: modulation of

CCL21. PLoS ONE 2010;5(1):e8955

101. Justo P, Sanz AB, Sanchez-Nino MD,

et al. Cytokine cooperation in renal

tubular cell injury: the role of TWEAK.

Kidney Int 2006;70(10):1750-8

102. Moreno JA, Izquierdo MC,

Sanchez-Nino MD, et al. The

inflammatory cytokines TWEAK and

TNFa reduce renal klotho expression

through NFkB. J Am Soc Nephrol

2011;22(7):1315-25.. Characterization of the molecular

mechanisms leading to Klotho

depletion in AKI and inflammation.

103. Izquierdo MC, Sanz AB,

Sanchez-Nino MD, et al. Acute kidney

injury transcriptomics unveils a

relationship between inflammation

and ageing. Nefrologia

2012;32(6):715-23

104. Izquierdo MC, Perez-Gomez MV,

Sanchez-Nino MD, et al. Klotho,

phosphate and inflammation/ageing in

chronic kidney disease.

Nephrol Dial Transplant

2012;27(Suppl 4):iv6-10. An updated summary of Klotho

physiology and its impact in kidney

injury and its consequences.

105. Macedo E, Bouchard J, Mehta RL. Renal

recovery following acute kidney injury.

Curr Opin Crit Care 2008;14(6):660-5

106. Forbes JM, Hewitson TD, Becker GJ,

et al. Ischemic acute renal failure:

long-term histology of cell and matrix

changes in the rat. Kidney Int

2000;57(6):2375-85

107. Ortiz A, Ucero AC, Egido J. Unravelling

fibrosis: two newcomers and an old foe.

Nephrol Dial Transplant

2010;25(11):3492-5. A didactic overview of the potential

clinical impact and mechanisms of

action of recently described mediators

of the transition from AKI to

kidney fibrosis.

108. Zeisberg M, Kalluri R. The role of

epithelial-to-mesenchymal transition in

renal fibrosis. J Mol Med (Berl)

2004;82(3):175-81

109. Jin Y, Ratnam K, Chuang PY, et al.

A systems approach identifies HIPK2 as

a key regulator of kidney fibrosis.

Nat Med 2012;18(4):580-8. Identification of a druggable kinase as

a potential therapeutic target for AKI

and kidney fibrosis.

110. Mazzinghi B, Ronconi E, Lazzeri E,

et al. Essential but differential role for

CXCR4 and CXCR7 in the therapeutic

homing of human renal progenitor cells.

J Exp Med 2008;205(2):479-90

111. Solomon A, Weiss DT, Kattine AA.

Nephrotoxic potential of Bence Jones

proteins. N Engl J Med

1991;324(26):1845-51

112. Ying WZ, Allen CE, Curtis LM, et al.

Mechanism and prevention of acute

kidney injury from cast nephropathy in a

rodent model. J Clin Invest

2012;122(5):1777-85. Description of a novel potential

therapy for myeloma cast nephropathy.

113. Faubel S, Chawla LS, Chertow GM,

et al. Ongoing clinical trials in AKI.

Clin J Am Soc Nephrol

2012;7(5):861-73.. An updated review of ongoing clinical

trials in AKI.

114. Cooper WA, O’Brien SM,

Thourani VH, et al. Impact of renal

A. Sanz et al.

16 Expert Opin. Drug Discov. [Early Online]

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.

Page 17: Progress in the development of animal models of acute kidney injury and its impact on drug discovery

dysfunction on outcomes of coronary

artery bypass surgery: results from the

Society of Thoracic Surgeons National

Adult Cardiac Database. Circulation

2006;113(8):1063-70

115. Loef BG, Epema AH, Smilde TD, et al.

Immediate postoperative renal function

deterioration in cardiac surgical patients

predicts in-hospital mortality and

long-term survival. J Am Soc Nephrol

2005;16(1):195-200

116. Fishbane S. N-acetylcysteine in the

prevention of contrast-induced

nephropathy. Clin J Am Soc Nephrol

2008;3(1):281-7

117. Chu-fan L, Xing W, Xun H, et al.

Protective effect of lipid microspheres

1 on myocardial injury following elective

percutaneous coronary intervention in

patients with angina pectoris: a pilot

study. J Cardiovasc Med (Hagerstown)

2011;12(11):790-4

118. Moore E, Bellomo R. Erythropoietin

(EPO) in acute kidney injury.

Ann Intensive Care 2011;1(1):3

119. Song YR, Lee T, You SJ, et al.

Prevention of acute kidney injury by

erythropoietin in patients undergoing

coronary artery bypass grafting: a pilot

study. Am J Nephrol 2009;30(3):253-60

120. Mitaka C, Kudo T, Haraguchi G, et al.

Cardiovascular and renal effects of

carperitide and nesiritide in cardiovascular

surgery patients: a systematic review and

meta-analysis. Crit Care 2011;15(5):R258

121. Ricci Z, Polito A, Ronco C. The

implications and management of septic

acute kidney injury. Nat Rev Nephrol

2011;7(4):218-25

122. Thompson JD, Kornbrust DJ, Foy JW,

et al. Toxicological and pharmacokinetic

properties of chemically modified siRNAs

targeting p53 RNA following intravenous

administration. Nucleic Acid Ther

2012;22(4):255-64

123. A dose escalation and safety study of

I5NP to prevent acute kidney injury

(AKI) in patients at high risk of AKI

undergoing major cardiovascular surgery

(QRK.004). ClinicalTrials.gov Identifier:

NCT00683553. Available at: http://

www.clinicaltrials.gov/ct2/show/

NCT00683553?term=I5NP&rank=2

[Last accessed 16 April 2013]

124. T€ogel F, Hu Z, Weiss K, et al.

Administered mesenchymal stem cells

protect against ischemic acute renal failure

through differentiation-independent

mechanisms. Am J Physiol Renal Physiol

2005;289(1):F31-42

125. A study to evaluate the safety and efficacy

of AC607 for the treatment of kidney

injury in cardiac surgery subjects (ACT-

AKI). ClinicalTrials.gov Identifier:

NCT01602328. Available at: http://www.

clinicaltrials.gov/ct2/show/NCT01602328?

term=ac607%2C+aki&rank=1 [Last

accessed 16 April 2013]

126. PGL5001 proof of concept study in

inflammatory endometriosis (JADE).

ClinicalTrials.gov Identifier:

NCT01630252. Available at: http://

www.clinicaltrials.gov/ct2/show/

NCT01630252?term=inflammatory

+disease%2C+jnk&rank=3 [Last accessed

16 April 2013]

127. Otani Y, Kijima T, Kohmo S, et al.

Suppression of metastases of small cell

lung cancer cells in mice by a peptidic

CXCR4 inhibitor TF14016. FEBS Lett

2012;586(20):3639-44

128. Besen A, Kose F, Paydas S, et al. The

effects of the nonsteroidal

anti-inflammatory drug diclofenac

sodium on the rat kidney, and alteration

by furosemide. Int Urol Nephrol

2009;41(4):919-26

129. Grollman AP. Aristolochic acid

nephropathy: harbinger of a global

iatrogenic disease. Environ Mol Mutagen

2013;54(1):1-7

130. Morishita Y, Ohnishi A, Watanabe M,

et al. Establishment of acute kidney

injury mouse model by 0.75% adenine

ingestion. Ren Fail 2011;33(10):1013-18

131. Nakamura A, Niimi R, Yanagawa Y.

Protection from sepsis-induced acute renal

failure by adenoviral-mediated gene transfer

of beta2-adrenoceptor.

Nephrol Dial Transplant 2010;25(3):730-7

132. Zhou X, Bao H, Takakura A, et al.

Polycystic kidney disease evaluation by

magnetic resonance imaging in

ischemia-reperfusion injured

PKD1 knockout mouse model:

comparison of T2-weighted FSE and

true-FISP. Invest Radiol 2010;45(1):24-8

133. Giraud S, Favreau F, Chatauret N, et al.

Contribution of large pig for renal

ischemia-reperfusion and transplantation

studies: the preclinical model.

J Biomed Biotechnol 2011;2011:532127

134. de Araujo M, Andrade L, Coimbra TM,

et al. Magnesium supplementation

combined with N-acetylcysteine protects

against postischemic acute renal failure.

J Am Soc Nephrol 2005;16(11):3339-49

135. Spandou E, Tsouchnikas I, Karkavelas G,

et al. Erythropoietin attenuates renal

injury in experimental acute renal failure

ischaemic/reperfusion model.

Nephrol Dial Transplant

2006;21(2):330-6

136. Clark LC, Farghaly H, Saba SR, et al.

Amelioration with vessel dilator of acute

tubular necrosis and renal failure

established for 2 days. Am J Physiol Heart

Circ Physiol 2000;278(5):H1555-64

137. Aravindan N, Natarajan M, Shaw AD.

Fenoldopam inhibits nuclear

translocation of nuclear factor kappa B in

a rat model of surgical ischemic acute

renal failure. J Cardiothorac Vasc Anesth

2006;20(2):179-86

138. Miller Q, Peyton BD, Cohn EJ, et al.

The effects of intraoperative fenoldopam

on renal blood flow and tubular function

following suprarenal aortic

cross-clamping. Ann Vasc Surg

2003;17(6):656-62

139. Molitoris BA, Dagher PC, Sandoval RM,

et al. siRNA targeted to p53 attenuates

ischemic and cisplatin-induced acute

kidney injury. J Am Soc Nephrol

2009;20(8):1754-64. Experimental basis for the clincial

trials of p53 siRNA in AKI.

140. Li Y, McLaren MC, McMartin KE, et al.

Involvement of urinary proteins in the

rat strain difference in sensitivity to

sensitivity to ethylene glycol-induced

renal toxicity. Am J Physiol

Renal Physiol 2010;299(3):F605-15

AffiliationAna B Sanz1,2 PhD,

Marıa Dolores Sanchez-Nino3 PhD,

Catalina Martın-Cleary1,2 MD,

Alberto Ortiz†1,2,4 MD PhD &

Adrian M Ramos1,2 PhD†Author for correspondence1Renal and Vascular Pathology Laboratory,

Instituto de Investigacion Sanitaria-Fundacio

Jimenez Dıaz/Universidad Autonoma de Madrid

(IIS-FJD-UAM), Madrid, Spain2Red de Investigacion Renal (REDINREN),

Madrid, Spain3IDIPAZ, Madrid, Spain4Fundacion Renal Inigo Alvarez de Toledo

(FRIAT), C/Jose Abascal, 42,

28003, Madrid, Spain

E-mail: [email protected]

Progress in the development of animal models of AKI and its impact on drug discovery

Expert Opin. Drug Discov. [Early Online] 17

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 05

/13/

13Fo

r pe

rson

al u

se o

nly.