16
Mechanisms of Tubulointerstitial Fibrosis Michael Zeisberg* and Eric G. Neilson *Division of Matrix Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; and Departments of Medicine and Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee Progressive tubulointerstitial fibrosis is the final common pathway for all kidney diseases leading to chronic renal fail- ure. 1,2 Its presence correlates with im- paired excretory function 1 and the de- gree of fibrosis 3,4 or fibroblast number 5 are robust pathologic markers of pro- gression. The histopathology of tubulo- interstitial fibrosis features deposition of interstitial matrix in association with in- flammatory cells, tubular cell loss, fibro- blast accumulation, and rarefaction of the peritubular microvasculature. 6 There is broad agreement that each of these hallmarks contributes to relentless pro- gression, although priority and interrela- tionship among the various components including their genetic predisposition are still muddled by unresolved com- plexity (Figure 1). Humans lose about 4500 nephrons per year per kidney, 7 and although not all agree, 8,9 classic inulin clearance studies suggest GFR falls by 10 ml/min per de- cade of life. 10 Therefore, the calculus for conceptualizing normal loss of renal function over time begins with a starting number of nephrons at birth integrated by the natural history of tissue involution from a lifetime of subclinical vascular disease, metabolic stress, cicatrization, and nephrosclerosis; 11–14 hence, the priv- ilege of aging provides a natural window into basic mechanisms of renal fibrogen- esis. Aging disturbs normal structure- function relationships in the kidney for many reasons, 15,16 including the influ- ence of telomere shortening on cell se- nescence, 16 –18 aberrant DNA methyl- ation destabilizing the epigenome of renal cells, 19 loss of self-renewing stem cells, 20 –22 and diminished rates of tubu- lar cell proliferation. 23 These nuclear consequences of aging are compounded by dysregulation of cellular energy sen- sors, 24 oxidative stress, 13,25,26 and mito- chondrial dysfunction 26,27 fogging the integrity of nephron structure with pro- gressive glomerular and tubulointersti- tial fibrosis. 12,13,28 PPAR agonists atten- uate some of these aging effects by engaging klotho, 29 stabilizing mitochon- drial deterioration, 29 and reducing the activity of TGF, 29,30 all to suggest that aging renal tissues are susceptible to the longitudinal affects of profibrotic sig- nals. 19,23,28,31 Persistent subclinical injury to the tubulointerstitium also affects the normal physiology of many nephrons at once, which explains the sensitivity of GFR to the spread of pernicious fibro- genesis. 1 Whereas the molecular mechanisms driving fibrogenesis are better under- stood from 20 years ago 1 and a variety of preclinical strategies to inhibit or even reverse tubulointerstitial fibrosis are ef- fective in rodents, only a few antifibrotic therapies are in clinical use today. 2,32–34 Finding better targets for future therapy in humans will require deeper under- standing of the molecular signals modu- lating fibrogenic events. THE EXTRACELLULAR MATRIX Excessive deposition of extracellular ma- trix, particularly the presence of collage- nous fibers, is the most striking and name-lending feature of tubulointersti- tial fibrosis. 6 By definition, fibrosis asso- ciates with both quantitative and qualita- Published online ahead of print. Publication date available at www.jasn.org. Correspondence: Dr. Michael Zeisberg, Harvard Medical School, Division of Matrix Biology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, RW 736C, Boston, MA 02215. Phone: 617-667-3583; Fax: 617-667-0360; E-mail: [email protected]; or Dr. Eric G. Neil- son, Departments of Medicine and Cell and De- velopmental Biology, D-3100 MCN, Vanderbilt University School of Medicine, Nashville, TN 37232-2358. Phone: 615-322-3146; Fax: 615-343- 9391; E-mail: [email protected] Copyright © 2010 by the American Society of Nephrology ABSTRACT The pathologic paradigm for renal progression is advancing tubulointerstitial fibrosis. Whereas mechanisms underlying fibrogenesis have grown in scope and understanding in recent decades, effective human treatment to directly halt or even reverse fibrosis remains elusive. Here, we examine key features mediating the molecular and cellular basis of tubulointerstitial fibrosis and highlight new insights that may lead to novel therapies. How to prevent chronic kidney disease from progressing to renal failure awaits even deeper biochemical understanding. J Am Soc Nephrol 21: 1819 –1834, 2010. doi: 10.1681/ASN.2010080793 BRIEF REVIEW www.jasn.org J Am Soc Nephrol 21: 1819–1834, 2010 ISSN : 1046-6673/2111-1819 1819

31. Mechanism of Tubulointerstitial Fibrosis

Embed Size (px)

DESCRIPTION

ckd

Citation preview

  • Mechanisms of Tubulointerstitial Fibrosis

    Michael Zeisberg* and Eric G. Neilson

    *Division of Matrix Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts;and Departments of Medicine and Cell and Developmental Biology, Vanderbilt University School of Medicine,Nashville, Tennessee

    Progressive tubulointerstitial fibrosis isthe final common pathway for all kidneydiseases leading to chronic renal fail-ure.1,2 Its presence correlates with im-paired excretory function1 and the de-gree of fibrosis3,4 or fibroblast number5

    are robust pathologic markers of pro-gression. The histopathology of tubulo-interstitial fibrosis features deposition ofinterstitial matrix in association with in-flammatory cells, tubular cell loss, fibro-blast accumulation, and rarefaction ofthe peritubularmicrovasculature.6 Thereis broad agreement that each of thesehallmarks contributes to relentless pro-gression, although priority and interrela-tionship among the various componentsincluding their genetic predispositionare still muddled by unresolved com-plexity (Figure 1).

    Humans lose about 4500 nephronsper year per kidney,7 and although not allagree,8,9 classic inulin clearance studiessuggest GFR falls by 10 ml/min per de-cade of life.10 Therefore, the calculus forconceptualizing normal loss of renalfunction over time begins with a startingnumber of nephrons at birth integratedby the natural history of tissue involution

    from a lifetime of subclinical vasculardisease, metabolic stress, cicatrization,andnephrosclerosis;1114 hence, the priv-ilege of aging provides a natural windowinto basic mechanisms of renal fibrogen-esis.

    Aging disturbs normal structure-function relationships in the kidney formany reasons,15,16 including the influ-ence of telomere shortening on cell se-nescence,1618 aberrant DNA methyl-ation destabilizing the epigenome ofrenal cells,19 loss of self-renewing stemcells,2022 and diminished rates of tubu-lar cell proliferation.23 These nuclearconsequences of aging are compoundedby dysregulation of cellular energy sen-sors,24 oxidative stress,13,25,26 and mito-chondrial dysfunction26,27 fogging theintegrity of nephron structure with pro-gressive glomerular and tubulointersti-tial fibrosis.12,13,28 PPAR agonists atten-uate some of these aging effects byengaging klotho,29 stabilizing mitochon-drial deterioration,29 and reducing theactivity of TGF,29,30 all to suggest thataging renal tissues are susceptible to thelongitudinal affects of profibrotic sig-nals.19,23,28,31 Persistent subclinical injury

    to the tubulointerstitium also affects thenormal physiology of many nephrons atonce, which explains the sensitivity ofGFR to the spread of pernicious fibro-genesis.1

    Whereas the molecular mechanismsdriving fibrogenesis are better under-stood from 20 years ago1 and a variety ofpreclinical strategies to inhibit or evenreverse tubulointerstitial fibrosis are ef-fective in rodents, only a few antifibrotictherapies are in clinical use today.2,3234

    Finding better targets for future therapyin humans will require deeper under-standing of the molecular signals modu-lating fibrogenic events.

    THE EXTRACELLULAR MATRIX

    Excessive deposition of extracellular ma-trix, particularly the presence of collage-nous fibers, is the most striking andname-lending feature of tubulointersti-tial fibrosis.6 By definition, fibrosis asso-ciates with both quantitative and qualita-

    Published online ahead of print. Publication dateavailable at www.jasn.org.

    Correspondence: Dr. Michael Zeisberg, HarvardMedical School, Division of Matrix Biology, BethIsrael Deaconess Medical Center, 330 BrooklineAvenue, RW 736C, Boston, MA 02215. Phone:617-667-3583; Fax: 617-667-0360; E-mail:[email protected]; or Dr. Eric G. Neil-son, Departments of Medicine and Cell and De-velopmental Biology, D-3100 MCN, VanderbiltUniversity School of Medicine, Nashville, TN37232-2358. Phone: 615-322-3146; Fax: 615-343-9391; E-mail: [email protected]

    Copyright 2010 by the American Society ofNephrology

    ABSTRACTThe pathologic paradigm for renal progression is advancing tubulointerstitialfibrosis. Whereas mechanisms underlying fibrogenesis have grown in scope andunderstanding in recent decades, effective human treatment to directly halt oreven reverse fibrosis remains elusive. Here, we examine key features mediating themolecular and cellular basis of tubulointerstitial fibrosis and highlight new insightsthat may lead to novel therapies. How to prevent chronic kidney disease fromprogressing to renal failure awaits even deeper biochemical understanding.

    J Am Soc Nephrol 21: 18191834, 2010. doi: 10.1681/ASN.2010080793

    BRIEF REVIEW www.jasn.org

    J Am Soc Nephrol 21: 18191834, 2010 ISSN : 1046-6673/2111-1819 1819

  • tive changes in matrix.1 In fibrotickidneys the widened interstitial spacesfill with fibrillar material consisting ofpredominantly collagens type I and IIIand fibronectin.1,35 This fibrotic matrixalso contains residual fragments of colla-gen type IV, which are normally found inbasement membranes otherwise sup-porting intact endothelia or tubular epi-thelia36,37 as well as several fibronectinsplice variants that modulate fibrogenicpotential.38 The actual amount of colla-gen produced by renal fibrogenesisseems to attenuate fairly soon after itstarts, whereas the relative extent of fi-brosis seems to increase with time.39 Thisdynamic incongruity can be reconciledby recognizing that the volume of in-jured kidney also decreases as residual re-nal parenchyma collapses around inva-sive interstitial scars.40

    Whereas fibrillar collagens spontane-ously assemble in vitro, this is not whathappens in vivo.41 During normal andabnormal tissue remodeling, interstitialcollagens have numerous binding part-ners and those critical for fibrillar assem-bly include fibronectin, collagen type V,fibronectin and collagen-binding inte-grins, and various fibrillins, including la-tent TGF-binding proteins (LTBP).42,43

    Prototypical matrix deposition follows a

    process where the initial appearance ofcollagen nucleators in interstitial fluidserve as scaffolding for fibrillar collagensproduced by local fibroblasts.43 Fi-bronectin, collagen type V, LTBP, andsecreted protein acidic and rich in cys-teine (SPARC) are interactive regulatorsof this matrix assembly:4244 fibronectinco-localizes with procollagen secretionto establish conformation-dependentrelationships with 51 integrins onfibroblasts43 and collagen type V oper-ates at the fibrillar core of collagen typeI assembly.45 LTBPs facilitate the secre-tion of pro-TGF and provide a mech-anism for its release and activation.42

    SPARC is antiproliferative,46 stimu-lates the expression of matrix metallo-proteinases (MMPs)47 and plasmino-gen activator inhibitor-1 (PAI-1),48

    and activates integrin-linked kinase(ILK),46 which engages epithelial-mes-enchymal (EMT) or endothelial-mes-enchymal transitions (EndMT) pro-ducing fibroblasts.49 51 All of theseprocesses shape the collagenous matrixfound in normal or disturbed tissues.

    Generally speaking, fibrogenesis ini-tiates in small areas at random sites ofinflammation and then expands to be-come diffuse if profibrotic drivers per-sist.52 The accumulation of fibroblasts in

    these damaged tissues is linked to the riskof fibrosis.5 Unfortunately, it is difficulttechnically to assign a definitive secre-tory role for collagen to any particularcell type found in these fibrogenic hotspots, and consequently, most in vivo ex-periments rely on surrogate markers ofcollagen secretion such as in situ hybrid-ization of mRNA encoding collagenchains,53 collagen promoter activity,54,55

    or the presence of collagen-containingcells.56 Better still, the endoplasmic ex-pression of the collagen type I chaperone,HSP47, seems more related to secre-tion,5760 and cell-specific deletions thatassociate with decreasedmatrix accumu-lation strongly insinuate secretory func-tion.61

    Fibroblasts use collagen fibrils as scaf-folding to crawl among damaged tissueplanes, and mathematical modeling sug-gests that fibroblast number and move-ment depends on their sensitivity to che-moattractant diffusion.62 These gradientsincrease collagen alignment in fibrogenictissue to facilitate the accrual of fibroblasts.The details of these events can be difficulttoassess in tissuesbiochemically, as impor-tant modulators of phenotypic change,particularly cytokines,mayexpress at levelsnot easily detectable, and in vitro activatorsor inhibitors may be artificially binary justby their presence in serum.63However, thepresence of PDGF and the absence ofTGF and FGF2/bFGF in fetal woundsthat do not scar compared with the pres-ence of all three cytokines in adult tissuethatdosuggestsdifferential expressionpat-terns of key growth factors may be neces-sary for enduring fibrogenesis.63 Prototyp-ical TGF signaling through Smadpathways64,65 under microRNA66 and epi-genetic control67,68 has one of the strongestprofibrotic effects on matrix produc-tion.69,70 The proximal action of angioten-sin II as a morphogenic cytokine stimulat-ing TGF71 and PAI-172 has also led to thewell-known renoprotective effects of re-nin-angiotensin inhibition.2,73 Whereascontainment of TGF signaling has beenthe centerpiece of various experimentalantifibrotic therapies,7480 knockdown ofTGF type II receptors along the collectingduct paradoxically accelerates renal fibro-sis associated with ureteral obstruction,81

    Figure 1. Interactive relationships producing fibrosis. Renal fibrosis constitutively in-volves inflammation, fibroblast activation, injury to the tubular epithelium, and micro-vascular rarefaction. Our understanding of how inflammatory cells, fibroblasts, tubularepithelial cells (TECs), and endothelial cells actively contribute to fibrogenesis hasevolved considerably during the past 20 years and sustained growth factor pressurewithin the microenvironment and increased susceptibility to growth factormediatedstimulation emerge as the principal driving force of fibrosis. The molecular systems thatdetermine choice between pathologic fibrosis and physiologic repair however are stillevolving. Epidemiologic studies identify genetic polymorphisms, epigenetic modifica-tions, and aging as risk factors for chronic kidney disease. Linking these risk factorsmechanistically to fibrosis is a task for the future.

    BRIEF REVIEW www.jasn.org

    1820 Journal of the American Society of Nephrology J Am Soc Nephrol 21: 18191834, 2010

  • all to suggest that too much or too little ofeven a single cytokine may be profibro-genic.82

    TISSUE PROTEASES

    Removal of extracellular matrix from theinterstitiumhingesonawell-wornbelief inthe role of various endogenous proteases.Most studies in this area focus on two fam-ilies of proteases: MMPs and members ofthe plasmin-dependent pathway. Bothclasses of proteases have the potential tofragment some extracellular matrix for re-moval, but also cleave nonmatrix sub-strates, releasing profibrotic growth factorsthat paradoxically trigger unwelcomeconsequences.

    MMPs are a family of zinc-dependentendopeptidases that currently includes25 members with varying substrate spec-ificities controlled by tissue inhibitors ofmetalloproteinases 1-4.83 The overlap-ping activity and specificity of MMPsmake it difficult to dissect individual ac-tions in vivo. Most studies in the kidneyfocus on MMP-2 and MMP-9 (bothdegrade collagen type IV84 and perhapscollagen types I and III85), but fail todemonstrate a consistent antifibroticeffect in the interstitium.86,87 For exam-ple, combined pharmacologic inhibitionof MMP-2, MMP-3, and MMP-9 at ad-vanced stages of disease in Alport mice ac-celerates renal fibrogenesis, whereas com-bined MMP inhibition before onset oftubulointerstitial fibrosis is protective.88

    More confusing, studies utilizing MMPnull mice show unaccelerated fibrogen-esis, and overexpression ofMMP-2 in tu-bular epithelial cells is sufficient to in-duce tubulointerstitial fibrosis.8789 Theprofibrotic effects of MMP-2 andMMP-9 on renal fibrosis, particularly theactivation of MMP-2 and MMP-14(MT1-MMP) by TGF associated withthe degradation of basement membranestimulating EMT,87 seem to outweightheir antifibrotic potential; in fact, micedeficient in TIMP-3 spontaneous de-velop interstitial fibrosis.90 MMP-14, amembrane-bound activator of secretedMMP-2 and MMP-9 in fibroblasts, iscritical for invasiveness into the intersti-

    tial environment containing collagentypes I and III85 and its expression is un-der the control of Snail, which is a keyregulator of the EMTprogram.91,92 Thus,epithelial cells, engaged by TGF, ex-press essential MMPs for both basementmembrane degradation and interstitialinvasion that are ultimately required forsuccessful completion of EMTor detach-ment and loss into the tubular lumen(Figure 2).

    The effects of the plasminogen-plas-min system on fibrosis are equally com-plex. Active plasmin is derived from pro-teolytic cleavage of plasminogen bytissue-type plasminogen activator (tPA)or urokinase-type plasminogen activator(uPA), which in turn are inhibited byPAI-1.93 Plasmin degrades some matrixconstituents, including laminin, entactin,perlecan, and fibronectin,93,94 whereasfibrillar collagen seems more resistant.95

    Plasmin, perhaps more importantly, alsoaffects cell behavior and function in thefibrotic environment and promotes fi-

    brogenesis by activating MMPs96 andstimulating EMT.97 tPA null mice withureteral obstruction have reduced localexpression of MMP-9 and preserved tu-bular basementmembrane with less EMT,and less interstitial fibrosis,98 whereasurokinase cellular receptor (uPAR) nullmice develop worse renal fibrosis withlower expression levels of the antifibroticcytokine, hepatocyte growth factor(HGF).99 Angiotensin II stimulates thePAI-1 promoter in tubular cells100 andtubulointerstitial fibrosis associates withTGF-stimulated PAI-1 expression,101

    whereas the genetic deletion of PAI-1ameliorates renal fibrogenesis inmice.72,102,103 The small heterodimerpartner (SHP) represses the transcrip-tion of TGF/Smad3regulated PAI-1expression, and SHP null mice developmore renal fibrosis, whereas SHP over-expression inhibits its development;101

    SHP may turn out to be an interestingtherapeutic candidate depending on itsmolecular specificity.

    Figure 2. Tissue proteases and tubular decondensation. Whereas proteases are keymodifiers of interstitial matrix, they also are critical for the disruption of basementmembrane. Tubular epithelial cells receive signals from the microenvironment tochange phenotype. As they release from basement membrane, they can either roundup and fall into the tubular lumen or undergo epithelial-mesenchymal transition andinvade the interstitium through rents in damaged basement membrane. SPARCthrough ILK, angiotensin II, and TGF activate nuclear programs (SHP, Snail1, andSmads) that engage in EMT-forming fibroblasts. Part of this mechanism is to stimulatethe release of PAI-1, MMP-2, and MMP-9. MMP-2 and MMP-9 are activated bymembrane-bound MMP-14, which is also modulated by PAI-1, tPA, and plasmin effectson MMPs.

    BRIEF REVIEWwww.jasn.org

    J Am Soc Nephrol 21: 18191834, 2010 Mechanisms of Tubulointerstitial Fibrosis 1821

  • The profibrotic action of these pro-teases is a paradigm shift in thinkingabout the dynamics of matrix depositionor its dissolution (Figure 2), illustratingthe complexity of balancing modifier ef-fects, timing and context, and weighingwhat seems to be true in vitrowithwhat isnot observed in vivo. Whereas currentevidence argues against MMPs or plas-min to degrade only complexmatrixes invivo, murine studies reporting regressionof tubulointerstitial fibrosis do demon-strate less matrix deposition.104107 Howthis matrix is removed biochemically, orfails to form, is still unclear. Involvementof other proteases, perhaps the lysosomalfamily of cysteine protease cathepsins,may be important.108112 The whole ap-proach to the regulation of collagen depo-sition in renal tissue is ripe for new insight.

    INFLAMMATORY CELLS

    Tubulointerstitial fibrosis is typically con-ditioned by the infiltration of inflamma-tory cells113 including dendritic cells,114

    lymphocytes,1,115,116 macrophages,117 andmast cells.118 Whereas inflammation as awhole contributes significantly to the en-gagement of fibrogenesis, evidence in re-cent years also highlights the antifibroticeffects of various subsets of lymphocytesand macrophages, providing novel po-tential avenues for antifibrotic therapies.

    Innate immunity through the activa-tion of toll-like receptors 4 (TLR4)119

    andTLR9,120 unlike TLR2 receptors,121 isan important early mediator of renal fi-brogenesis. Lymphocytes precede the in-flux of macrophages.1 Rag-2 null mice,which lack bothmature B andT lympho-cytes, are protected from fibrosis, dem-onstrating that effector lymphocytes arean essential early component of fibro-genesis.116 Adoptive transfer of CD4

    cells into Rag/ mice, but not CD8

    cells so much, increases fibrogenesis.122

    The contribution of CD3 T cells to re-nal fibrogenesis is well established, as ac-tivated cytotoxic T cells attack tubularepithelial cells.2 Administration of PGE1inhibits the effector arm of T lympho-cytes in interstitial nephritis that is over-come by competitive exposure to IL-1.123

    Long-term renal fibrosis after ischemia-reperfusion injury also depends on per-sistent infiltration of effector-memory Tlymphocytes, which correlate with onsetof fibrosis in later stages.124 Transfer of Tlymphocytes from mice with prolongedischemia-reperfusion injury to nave re-cipients is sufficient to induce renal in-jury, further highlighting the relevance ofmemory lymphocytes in theprogressionoftubulointerstitial fibrosis.125 In contrast,FoxP3 regulatory T cells (Tregs) bluntkidney injury and are required for physi-ologic kidney regeneration.126 Whereasrecent studies suggest the presence ofCD20B cells are as common asCD3Tlymphocytes in established fibrosis, thefunctional contribution of B cells to renalfibrogenesis is uncertain.2,127,128

    When monocytes are recruited todamage the interstitium, they transdif-ferentiate into macrophages.129 There isa strong correlation between macro-phage infiltration and the extent of fibro-sis.130 Activation of TRL9 on macro-phages by CpG-oligodeoxynucleotides isan accelerant for interstitial fibrogen-esis,120 suggesting the dispersion of nu-clear fragments after local cell death mayenhance inflammation. Depletion ofmacrophages also ameliorates fibrogen-esis in mice, highlighting the traditionalprofibrotic view of macrophages.131,132

    Macrophages however can be catego-rized into classically activated M1 mac-rophages or alternatively activated M2macrophages. Unlike the M1 macro-phages, M2 macrophages are anti-in-flammatory and provide cues for tissuerepair.129 Infusion of cells enriched forM2 macrophages ameliorates renal fi-brosis in mice.133 Finally, in addition tomacrophages, dendritic cells are critical toantigen processing in tubulointerstitial in-jury;114 their proteasomal processing of al-bumin, for example, creates new antigenictargets.134 The balance or relative presenceof these subsets of cells in renal inflamma-tion over time needs more study.

    Mast cells are well-established con-tributors to fibrosis in lung, liver, andpancreas where they release proinflam-matory cytokines, including chemokinesCCL2-5, TNF, and leukotrienes.118

    Surprisingly, however, mast celldefi-

    cient KitW-sh/KitW-sh mice display en-hanced fibrosis in the kidney.118,135

    Such accelerated fibrosis associateswith increased levels of TGF, suggest-ing that mast cells in kidney play a rolein suppressing the fibrotic process.118

    Several new biologic modifiers of theimmune system reduce interstitial fibro-sis. PPAR agonists reduce the numberof macrophages by attenuating TGFexpression,30 and three independentstudies report the administration of aCCR1 antagonist, anti-TNF blockingantibodies, or an IL-1 receptor antago-nist are sufficient to ameliorate renalfibrosis, even when treatment is initi-ated in advancing disease.136138 Thesetherapeutic studies corroborate therole of inflammation not only in theinitiation but also in perpetuation offibrogenesis.

    FIBROBLASTS

    Fibroblasts in the renal interstitium areconsidered the principal source of fibril-lar matrix (collagen types I and III), andtubulointerstitial fibrosis inevitably asso-ciates with a robust accumulation of fi-broblasts,5 displaying increased intrinsicproliferative activity139,140 or, in some fi-broblasts, what is called an activated phe-notype expressing -smooth muscle ac-tin (SMA).70,141,142 Tissue fibroblastsexhibit phenotypic heterogeneity,143145

    more likely based on origin,54,146,147 butthis heterogeneity in tissue is also bimo-dal: those cells that are RhoA-dependentSMA148 and those that are not. Onegeneric dilemma with SMA is thatwhen used as a fibroblast marker in in-jury, it is not distinguishable fromSMA mural cellsvascular smoothmuscle cells or venular pericytes;149,150

    nor does thismarker detect the larger popu-lation of SMA fibroblasts.58,146,147,151,152

    The view of myofibroblasts as principal me-diators of renal fibrosis is also based on thesuggestion that fibrosis associates withde novo accumulation of SMA

    cells.51,153,154 This notion, however, hasto be tempered by observations thatSMA fibroblasts do notmove,155 renalfibrogenesis persists despite decreasing

    BRIEF REVIEW www.jasn.org

    1822 Journal of the American Society of Nephrology J Am Soc Nephrol 21: 18191834, 2010

  • numbers of SMA myofibroblasts,121

    SMA fibroblasts contain and likelyexpress interstitial collagens in vivo,61,152

    and mice deficient of SMA developmore fibrosis and their fibroblasts pro-duce more collagen type I than SMA

    fibroblasts.156 Unequivocal functionaldata to support the idea that nonmotileSMA myofibroblasts principally me-diate fibrogenesis is lacking.

    The most durable fibroblast markerin the kidney is fibroblast-specific pro-tein-1 (FSP1; S100A4).58,146,147,151 Theproclivity for FSP1 in fibroblasts, amem-ber of the S100 family of calcium-bind-ing proteins, was discovered in a differ-ential genetic screen between fibroblastsand epithelial cells,157 and renal fibrosisassociates with a robust accumulation ofFSP1 fibroblasts.5,61,157 A few studiessuggest FSP1 is expressed by macro-phages;55,158,159 however, this notion per-sists from studies using underdetectionmethods for FSP1/S100A4 that may en-hance nonspecific staining and/or fromimproper application of so-called anti-macrophage antibodies that share targetspecificities with fibroblasts and are notmacrophage-specific;158160 FSP1 pro-moter activity in fibroblasts driving aGFP reporter60,161 or Cre recombinase162

    clearly discriminate tissue fibroblasts fromF4/80macrophages.FSP1, F4/80 fibro-blasts are also the only population in thekidney proven to contribute to fibrogen-esis, as their ablation in FSP1-tk trans-genic mice ameliorates fibrosis substan-tially.61 This kind of functional deletionexperiment is critical evidence not yet re-ported in FSP1 cells expressing SMA,particularly pericytes.54 For themoment,it is best to think of all tissue fibroblasts aspotentially fibrogenic.58,147,152

    The true heterogeneity of fibroblastsin the kidney may be understood by ex-amining the different cellular origins ofthese cells.147 Pools of fibrogenic fibro-blasts comingle either by proliferation ofresident fibroblasts,163 some of whichmay be subject to local or time-depen-dent epigenetic effects;68,164 from localtubular epithelial cells51,59,157,165 or endo-thelial cells (simple squamous epithe-lia)51,166 after EMT or EndMTduring tis-sue injury; after recruitment of FSP1,

    CD34 bone marrowderived fibro-cytes;59,146 and perhaps fromblood vesselshedding of SMA pericytes54,150 thatderive developmentally from mesothe-lium through EMT.167,168 It is not yetclear whether FSP1, SMA peri-cytes54,55 should be considered true fi-broblasts or a separate and unique phe-notype of collagen-producing cells.150 Asopposed to tissue-derived fibroblasts,CD34 bone marrowderived fibro-cytes59 may also have a unique lineagefromCD11b, CD115, Gr1 leukocyteprogenitors under the control of CD4Tcells;56 some studies suggest these circu-lating fibrocytes do not contribute to tis-sue fibrogenesis,53,55 whereas other ex-periments suggest they can to a limitedextent.50,56,59 For the moment, it isprobably best to view multiple lineagesas contributing to the final mix of fi-broblast populations in tissue. It is en-tirely unknown if these various lin-eages have priority among individualsor different diseases or if there is indi-vidual variation or preference as fibro-genesis progresses.

    Depletion of interstitial fibroblasts byselectively corrupting their DNA replica-tion is one classic strategy for experimen-tal antifibrotic therapy.61 Conditionalexpression of an IB dominant-negativetransgene in fibroblasts also attenuatesfibrogenesis by inhibiting NF-B.60 Butbecause specific targeting of fibroblastshas not yet been achieved without ge-netic manipulation, current antifi-brotic therapies center on variousother approaches; these approaches in-clude blocking PDGF-C,169 PDGF-D,170 or TGF171,172 with neutralizingantibodies or using pirfenidone,173175

    pyridoxamine,176 ALK5 receptor ki-nase inhibitors,177 paricalcitol,178 Rasinhibitors,68,179 or PPAR agonists.30

    Blockade of AT1R by the small moleculeinhibitorCGP-48933 also attenuates inter-stitial fibrosis,180 as does inhibition of an-giotensin II73 or aldosterone action.32,33

    TUBULAR EPITHELIUM

    Tubular epithelial cells spark the pro-gression of fibrogenesis to their own

    detriment. They do this by modulatingtheir proliferation,23,181 or by provid-ing proinflammatory chemokines andcytokines that induce mononuclear in-flammation and the formation of fi-broblasts (Figure 3).59,182184 These localcytokines provide critical signals mediat-ing primary interstitial injury.185 Sec-ondary interstitial damage after glomer-ulonephritis also depends on the tubulartoxicity of glomerular proteinuria,1,186

    the filtration of protein-bound cytokinesfrom plasma to the tubular microenvi-ronment,184,187 or possible cytokine leak-age through bulk fluid flow from the af-ferent arteriole directly into the interstitialspaces associated with the juxtaglomerularapparatus.188 Hydrodynamic forces alongthe tubules themselves are also profi-brotic.189Tubular stretch fromobstructioninduces TGF and NF-B; increases insinglenephronGFRandcompensatory in-creases in fluid shear stress also decreasetPA and increase TGF.

    Theepicenter forearlymolecularactionby tubular epithelial cells is through NF-B.190 Tubular cells toggle on NF-Bafter exposure to proteins from tubularfluid,134,191 activation of CD36 scavengerreceptors,25 or exposure to connective tis-sue growth factor,192 CD40 ligand,193 an-giotensin II,194,195 or aldosterone.32,33 Acti-vation ofNF-B is critical for initiating thedownstream release of PAI-1 or chemo-kines and growth factors,196 particularlyIL-1,137 IL-6,197MCP-1/CCL2,198,199RAN-TES/CCL5,113,200 or TNF.200 In opposi-tion, administration of Smad7,201,202 pari-calcitol,203 truncated IB,204 HGF,200,205

    spironolactone,32,206 and decoyNF-Boli-godeoxynucleotides207 all attenuate tubu-lointerstitial injury by inhibiting the activ-ity of NF-B. Experimental inhibition ofproinflammatory chemokines such asTNF or CCR1 has also been successfulin preventing progressive interstitialinjury.113,138

    With time, persistent tubulointersti-tial injury inevitably associates with mi-tochondrial dysfunction or loss,26,208 for-mation of aglomerular tubuli,209 tubularatrophy,27 and rampant interstitial fibro-sis,146 creating the classic pathologicpicture of renal progression.210 Majorepithelial mechanisms contributing to

    BRIEF REVIEWwww.jasn.org

    J Am Soc Nephrol 21: 18191834, 2010 Mechanisms of Tubulointerstitial Fibrosis 1823

  • chronic tubulointerstitial fibrosis areG2/M cell cycle arrest,181 cellular apo-ptosis,211 and EMT-forming fibro-blasts;147,151 all three processes aremodulated by the transcriptional re-pressor GLIS2.212 EMT is a fundamentalprocess to create cells that will move,155 amodification in lineage maturation thatdisturbs the stateofnucleardiapause in ep-ithelial andendothelial cells.59,213Althoughnot all agree,54,159,214,215 11 studies confirmthese transitions to fibroblasts in a varietyof lineage-tracing experiments fromdiffer-ent tissues, including kidney,50,59,60,165

    intestine,216 liver,217 heart,166 lung,218220

    and endothelium50,166,221 using 10 differ-ent cell fate reporter constructs.

    The role of EMT in forming renal fi-broblasts has been recently reviewed49

    and debated;160 several of the lineage-tracing studies unable to detect EMT orFSP1 fibroblasts are likely confoundedbyuse of nonpreferred anti-FSP1/S100A4 an-tibodies and/or inadequate antigen-re-trieval methods during tissue prepara-tion,54,159,215 by relying on a collagenpromoter driving green fluorescent pro-tein used to detect fibroblasts that per-haps may not express in all fibroblastsbecause of methylation or enhancer bi-as,160 and by overlooking lineage evi-dence that SMA mural cells/peri-

    cytes may have emerged previouslyfrom EMT.167,168,222 It is increasinglyclear that fibroblasts derive from mul-tiple parental lineages, including epithe-lial cells, endothelial cells, and bonemar-row endosteal cells. The status ofpericytes in fibrogenesis remains an areaof great interest that will require morefunctional evidence of their role one wayor the other. Finally, although EMT isdifficult to assess in humans who are notbiopsied until there is clinical evidence ofdisease,5 early renal allograft biopsiessuggest changes in epithelial phenotypesupportive of transitions preceding fi-brosis,223225 although not all agree.226

    Glis2 mutations producing autosomalrecessive NPHP7, a rare form ofnephronophthisis, may be the first sug-gestive evidence of spontaneous or dere-pressed EMT causing renal fibrosis inhumans.212,227

    EMT induces a variety of intermedi-ate cell phenotypes, not all of which com-plete their transition to fibroblasts.147,225

    This intermediacy and its reversibilitymay depend on the variable persiste-nce of endogenous perturbagens: hypo-xia,165,228230 TGF,231 EGF,231,232 PTH-RP,233 and plasmin97 all induce EMT, asdoes activation of ILK49 or RAGE.234 Vi-tamin D receptors downregulate the re-

    nin-angiotensin system driving EMTand TGF and receptor absence acceler-ates fibrogenesis.235

    After induction, a hierarchy of tran-scription factors program EMT, includ-ingGLIS2212 andCBF-A,236 amaster reg-ulator of Snail,237 Twist,238 HMGA2,LEF-1, and Ets-1.236 Overexpression ofSnail1237 induces EMT and fibrosis andrecessive mutations in Glis2 spontane-ously invoke EMT in tubular cells inmice with renal fibrosis;212 short inter-fering RNA knockdown of Snail1 alsoblocks EMT during vascular mural celldifferentiation.222 There appears to be agradualness to EMT events in the kid-ney,225 as indicated by the variably activesignaling of -catenin, Snail, or Twist inwatershed epithelial cells along chroni-cally injured tubules that is based on thetime-dependent permissiveness of localcytokine action. Whereas activation ofthe EMTprogram is ameans for parentalcells to resist proapoptotic stimuli,211,239

    such tenuous protection from cell deathalso results in loss of parental cell pheno-type.59,240 The true frequency of tubularepithelial cells that complete EMT andbecome fibroblasts is unknown, butlikely depends on the persistence of in-flammation, epigenetic control of fi-brotic gene regulation,67 and ultimatelythe emergence of tubular atrophy withacellular scars that are the consequenceof profibrotic cytokine exhaustion andtubular cell disappearance.

    The relevance of EMT for progressionof renal fibrosis is highlighted by studies inwhich administration of bonemorphogenicprotein-7 (BMP7),80 HGF,241,242 the small-molecule inhibitor ILK, QLT-0267,243 orY-27632,aninhibitorofRho/ROCK244all in-hibit tubular EMT and ameliorate fibrogen-esis. Trps1 operates downstream ofBMP7 in the kidney,245 and the availabilityof Smad7controlling thephosphorylationofSmad3 is normally assured by Trps1 inhibi-tion of ubiquintination through Arkadia;246

    Trps1 haploinsufficiency raises the levels ofArkadia,decreasingtheavailabilityofSmad7,and thus facilitating fibrosis through TGF/Smad3mediated EMT. Induction of heatshock protein-72 (HSP72) also protects tu-bular epithelia from apoptosis, EMT, and fi-brosis,239 and overexpression of klotho247 or

    Figure 3. The cytokine milieu in fibrogenesis. NF-B activators and inhibitors competefor the engagement of mammalian target of rapamycin and release of chemokines andproinflammatory cytokines that draw macrophages, dendritic cells, and T lymphocytesto the tubulointerstitium. These mononuclear cells injure the tubular epithelia and activatefibroblasts. The transcriptional activation of EMT and EndMT is engaged by hypoxia andvarious cytokines, particularly TGF, EGF, and ILK. EMT inhibitors block the developmentof fibroblasts that deposit extracellular matrix into the fibrogenic interstitium.

    BRIEF REVIEW www.jasn.org

    1824 Journal of the American Society of Nephrology J Am Soc Nephrol 21: 18191834, 2010

  • inhibition of themammalian target of rapa-mycin248 attenuates fibrogenesis in a varietyof experimentalmodels.

    Apivotal determinant in the failure to re-coverfrominterstitial injuryistherelativeab-sence of nephron regeneration in kidneysthatbecomefibrotic.181Onereasonfor this isthat glomerulotubular disconnections limitrepair of nephrons.107,209 The glomerulotu-bularneck at the antivascular pole of the glo-merular tuft is an at-risk break point for theformation of aglomerular tubuli249 and sur-prisingly angiotensin inhibition protectsthese unwanted separations.250 Another rea-son is that cell cycle arrest of tubular epithe-lium during acute kidney injurymay stimu-lateprofibrotic signaling.181Whereas tubularcells along atrophic tubules variably increasetheir proliferative activity in advanced tubu-lointerstitialdisease,210 this restorativepoten-tialmaydeclinewithagingthroughthemod-ulation of the adipokine, zinc-2-glycoprotein, knownasZag.23

    The regenerative capacity of injured tu-bules absent persistent inflammation is likelythe result of self-renewing tubular cells orperhaps progenitor cells within other nichesalong the nephron.31 A CD24, CD133

    progenitor cell niche has been identified inthe parietal epitheliumof Bowmans capsulenear the vascular pole,21,22,251 and the adop-tive transfer of these cells at the timeof tubu-lar injury obviates the appearance of intersti-tial fibrosis.252 The glomerulotubular neckregionisalsowhereexvivodifferentiatedem-bryonic stemcells traffic after adoptive trans-fer during development253 andmay be a tu-bular niche not unlike the vascular pole.21

    Expansion of renal progenitor cells is underthe control of histone deacetylases and theirenzymatic inhibition may offer restorativepotential to the kidney.254

    THE MICROVASCULATURE

    Rarefaction of the peritubular vascula-ture with ensuing chronic hypoxia arehallmarks of tubulointerstitial fibro-sis.107 In early stages of injury the peritu-bular vasculature is damaged by proapop-totic stimuli, particularly transientischemia.255,256Progressive fibrosis thenremodels the peritubular endothelialnetwork governed by imbalance among

    pro- and antiangiogenic stimuli257,258 orloss of peritubular endothelial cells byEndMT-forming fibroblasts.50,259

    The fibrotic tubulointerstitium is a hy-poxic environment.260,261 Hypoxia resultsfrommicrovascular rarefaction made worsebymatrix expansion throughout the intersti-tium,258 requiring theoxygengradient todif-fuse greater distances from vasoconstrictedor attenuated microvessels to adjacent isch-emic tubules.230,260 Hypoxia also contributesdirectly to the progression of tubulointersti-tial fibrosis by simulating EMTandpromot-ing matrix accumulation from new fibro-blasts.165

    Different strategies to improve thischronic hypoxia during renal progres-sion show some promise in experimentalmodels of renal fibrosis. Angiotensin IIinhibition increases capillary blood flowand cortical oxygen levels in normal re-nal tissues,262 and long-acting calciumchannel blockers attenuates the hypoxiaof angiotensin IIinduced tubulointer-stitial injury.263 Inhibition of the vaso-constrictor, endothelin, is also renopro-tective,264 particularly in combinationwith ACE inhibition.265 Activation of thehypoxia-inducible factor (HIF) pathwayor stabilization of HIF proteins attenu-ates renal fibrosis aswell.230,261,266 The ul-timate goal of reversing microvascularrarefaction, however, has been difficultto achieve. Whereas the HIF pathway isan important inducer of angiogenic vas-cular endothelial growth factor (VEGF)proteins,230 systemic administration ofVEGF121 does not improve renal micro-vascular disease.267 Whereas administra-tion of the antifibrotic molecule, BMP7,blocks EMT in the kidney,80 its affect onEndMT in the kidney is unknown; how-ever, BMP7 does inhibit EndMT, endo-thelial loss, and fibrogenesis in experi-mental cardiac fibrosis.166

    GENETIC PREDISPOSITION TORENAL PROGRESSION

    Whereas revelations regarding new cellularmechanismsofrenalfibrogenesiscontinuetoflourish, the identification of key risk factorsunderlying the true biologic fate of renal tis-sue is far from clear. Risk factors for chronic

    kidneydiseaseare rapidly emerging fromep-idemiologic studies, including age,268,269

    nephronmass,7,270 gender,268,271 levels of cal-cidiol,272 sympathetic activity,273 obesity anddiabetes,274 cardiovascular disease,275 geneticloci276 and polymorphisms,273,277279 andepigenetic modifications.19 But how theserisk factors modulate fibrogenesis is still un-certain.

    Genetic polymorphisms in cytokine riskprofiles in some instances can be explainedusingexistingknowledgeof fibrotic signalingpathways.278 In other cases, it is unclearwhy genetic polymorphisms in uro-modulin280,281 or methenyltetrahydro-folate synthetase282 increase suscepti-bility to fibrogenesis. Insights into therole of epigenetics in renal progressionand fibrogenesis are also just starting toemerge. For example, aberrant hyper-methylation of selected genes includingRASAL1, a suppressor of Ras signaling,contributes to perpetual fibroblast acti-vation and fibrogenesis.68 Furthermore,there are large families of small, naturallyoccurring, noncoding RNAs that inter-fere post-transcriptionally with the stabil-ity or translation of coding mRNA;283 threeprototypical families of interfering RNAsinclude short interfering RNAs (siRNA),microRNAs (miRNAs), and piwi-interact-ing RNAs (piRNA).284 miRNA-192, forexample, promotes the activation ofTGF/Smad3-mediated fibrosis in ure-teral obstruction and 5/6 nephrec-tomy,285 and in diabetic glomeruli,TGF stimulates miRNA-192 to syner-gize with deltaEF1 short hairpin RNAsthat increase Col1a2 E-box activity inmesangial cells.286 Oddly enough, lowlevels of miRNA-192 in human kidneybiopsies of diabetic nephropathy cor-relate with late presentation and less fi-brosis.164 Thus, the interfering RNAstory becomes complicated quickly,largely because the network effect ofmultiple families is not addressed bystudying the action of a single familymember.

    CONCLUSIONS

    Major progress has been made duringthe last 20 years in characterizing the cel-

    BRIEF REVIEWwww.jasn.org

    J Am Soc Nephrol 21: 18191834, 2010 Mechanisms of Tubulointerstitial Fibrosis 1825

  • lular and molecular mechanisms of in-terstitial injury. The molecular eventsdetermining choice between self-limitedrepair of the tubulointerstitium or pro-gressive fibrogenesis suggest that persis-tent cytokine pressure and the accumu-lation of unwanted inflammatory cellsare the principal effectors of chronicity.There remains a gap in understandingthe genetic drivers of this cytokine pres-sure or sensitivity, and our knowledge ofhow genetic polymorphisms and epige-netic modifications facilitate fibrogen-esis are in their infancy.

    Modulating the pivotal role of tubularepithelial cells in their provision of pro-fibrotic stimuli, and the difficulty in pre-venting microvascular rarefaction, re-main new challenges for the future. Therole of determining cells, particularlymyofibroblasts or pericytes, in renal fi-brogenesis needs more clarification incomparison to the known functionalityof FSP1 fibroblasts. A deeper under-standing of the molecular basis for fibro-blast expansion and collagen accumula-tion also requires more work. Newtherapies to abrogate EMT are on the ho-rizon but the chromatin biology modu-lating these phenotypic changes are un-clear; determining the point of no returnfor intermediate states of transition isstill an open question.

    Today, the distinction between profi-brotic and antifibrotic stimuli also seemsincreasingly muddledsome macro-phage, T cell subpopulations, and mastcells, many of which were once viewed asunequivocally pernicious, now seemrenoprotective. Tissue proteases, alsoonce considered renoprotective, inmanyrespects now look as if they encouragefibrogenesis. The role of progenitor cellsin regeneration and the depletion of stemcell niches along the adult nephron isalso an underdeveloped area of investi-gation.

    Most importantly, although the pres-ence of tubulointerstitial fibrosis has al-ways implied an unalterable fate, thisperception is changing in new preclinicalstudies using unique biologic modifiers.We have renewed optimism that in thenot too distant future some of thesenovel insights will become clinically rel-

    evant for patients with chronic kidneydisease.

    ACKNOWLEDGMENTS

    Some of the cited work came from NIH

    grants DK-46282 (E.G.N.) and DK-81576,

    DK-81687, and DK-74558 (M.Z.). We thank

    Youhua Liu, Raymond Harris, Robert

    Colvin, and Yash Kanwar for helpful com-

    ments regarding earlier drafts of this manu-

    script, and we apologize for the lack of space

    to cite many other interesting findings.

    DISCLOSURESNone.

    REFERENCES

    1. Kuncio GS, Neilson EG, Haverty T: Mech-anisms of tubulointerstitial fibrosis. KidneyInt 39: 550556, 1991

    2. Harris RC, Neilson EG: Toward a unifiedtheory of renal progression. Annu Rev Med57: 365380, 2006

    3. Schainuck LI, Striker GE, Cutler RE, BendittEP: Structural-functional correlations in re-nal disease. II. The correlations. HumPathol 1: 631641, 1970

    4. Bohle A, Bader R, Grund KE, Mackensen S,Neunhoeffer: Serum creatinine concentra-tion and renal interstitial volume. Analysisof correlations in endocapillary (acute) glo-merulonephritis and in moderately severemesangioproliferative glomerulonephritis.Virchows Arch A Pathol Anat Histol 375:8796, 1977

    5. Nishitani Y, Iwano M, Yamaguchi Y, HaradaK, Nakatani K, Akai Y, Nishino T, Shiiki H,Kanauchi M, Saito Y, Neilson EG: Fibro-blast-specific protein 1 is a specific prog-nostic marker for renal survival in patientswith IgAN. Kidney Int 68: 10781085, 2005

    6. Bohle A, Christ H, Grund KE, Mackensen S:The role of the interstitium of the renalcortex in renal disease. Contrib Nephrol16: 109114, 1979

    7. Luyckx VA, Brenner BM: The clinical impor-tance of nephron mass. J Am Soc Nephrol21: 898910, 2010

    8. Fliser D, Franek E, Joest M, Block S, Mut-schler E, Ritz E: Renal function in the el-derly: Impact of hypertension and cardiacfunction. Kidney Int 51: 11961204, 1997

    9. Glassock RJ, Winearls C: Ageing and theglomerular filtration rate: Truths and con-sequences. Trans Am Clin Climatol Assoc120: 419428, 2009

    10. Davies DF, Shock NW: Age changes in glo-merular filtration rate, effective renalplasma flow, and tubular excretory capacityin adult males. J Clin Invest 29: 496507,1950

    11. Abrass CK, Adcox MJ, Raugi GJ: Aging-associated changes in renal extracellularmatrix. Am J Pathol 146: 742752, 1995

    12. Baylis C, Corman B: The aging kidney: In-sights from experimental studies. J Am SocNephrol 9: 699709, 1998

    13. Zhou XJ, Rakheja D, Yu X, Saxena R, VaziriND, Silva FG: The aging kidney. Kidney Int74: 710720, 2008

    14. Rule AD, Am H, Cornell LD, Taler SJ, CosioFG, Kremers WK, Textor SC, Stegall MD:The association between age and nephro-sclerosis on renal biopsy among healthyadults. Ann Intern Med 152: 561567, 2010

    15. Anderson S, Halter JB, Hazzard WR, Him-melfarb J, Horne FM, Kaysen GA, KusekJW, Nayfield SG, Schmader K, Tian Y, Ash-worth JR, Clayton CP, Parker RP, Tarver ED,Woolard NF, High KP: Prediction, progres-sion, and outcomes of chronic kidney dis-ease in older adults. J Am Soc Nephrol 20:11991209, 2009

    16. Yang H, Fogo AB: Cell senescence in theaging kidney. J Am Soc Nephrol 21: 14361439, 2010

    17. Melk A, Ramassar V, Helms LM, Moore R,Rayner D, Solez K, Halloran PF: Telomereshortening in kidneys with age. J Am SocNephrol 11: 444453, 2000

    18. Wills LP, Schnellmann RG: Telomere short-ening and regenerative capacity afteracute kidney injury. J Am Soc Nephrol 21:202204, 2010

    19. Dressler GR: Epigenetics, development,and the kidney. J Am Soc Nephrol 19:20602067, 2008

    20. Gupta S, Verfaillie C, Chmielewski D, KrenS, Eidman K, Connaire J, Heremans Y, LundT, Blackstad M, Jiang Y, Luttun A, Rosen-berg ME: Isolation and characterization ofkidney-derived stem cells. J Am Soc Neph-rol 17: 30283040, 2006

    21. Lasagni L, Romagnani P: Glomerular epi-thelial stem cells: The good, the bad, andthe ugly. J Am Soc Nephrol 2010, in press

    22. Romagnani P, Kalluri R: Possible mecha-nisms of kidney repair. Fibrogenesis TissueRepair 2: 3, 2009

    23. Schmitt R, Marlier A, Cantley LG: Zag ex-pression during aging suppresses prolifer-ation after kidney injury. J Am Soc Nephrol19: 23752383, 2008

    24. Ix JH, Sharma K: Mechanisms linking obe-sity, chronic kidney disease, and fatty liverdisease: The roles of fetuin-A, adiponectin,and AMPK. J Am Soc Nephrol 21: 406412, 2010

    25. Okamura DM, Pennathur S, Pasichnyk K,Lopez-Guisa JM, Collins S, Febbraio M,Heinecke J, Eddy AA: CD36 regulates ox-idative stress and inflammation in hyper-

    BRIEF REVIEW www.jasn.org

    1826 Journal of the American Society of Nephrology J Am Soc Nephrol 21: 18191834, 2010

  • cholesterolemic CKD. J Am Soc Nephrol20: 495505, 2009

    26. Coughlan MT, Thorburn DR, Penfold SA,Laskowski A, Harcourt BE, Sourris KC, TanAL, Fukami K, Thallas-Bonke V, NawrothPP, Brownlee M, Bierhaus A, Cooper ME,Forbes JM: RAGE-induced cytosolic ROSpromote mitochondrial superoxide gener-ation in diabetes. J Am Soc Nephrol 20:742752, 2009

    27. Moller JC, Skriver E, Olsen S,Maunsbach AB:Ultrastructural analysis of human proximal tu-bules and cortical interstitium in chronic renaldisease. Virchows Arch A Pathol Anat His-topathol 402: 209237, 1984

    28. Thomas SE, Anderson S, Gordon KL,Oyama TT, Shankland SJ, Johnson RJ: Tu-bulointerstitial disease in aging: Evidencefor underlying peritubular capillary dam-age, a potential role for renal ischemia.J Am Soc Nephrol 9: 231242, 1998

    29. Yang HC, Deleuze S, Zuo Y, Potthoff SA,Ma LJ, Fogo AB: The PPARgamma ago-nist pioglitazone ameliorates aging-related progressive renal injury. J Am SocNephrol 20: 23802388, 2009

    30. Kawai T, Masaki T, Doi S, Arakawa T,Yokoyama Y, Doi T, Kohno N, Yorioka N:PPAR-gamma agonist attenuates renal in-terstitial fibrosis and inflammation throughreduction of TGF-beta. Lab Invest 89: 4758, 2009

    31. Humphreys BD, Valerius MT, KobayashiA, Mugford JW, Soeung S, Duffield JS,McMahon AP, Bonventre JV: Intrinsic ep-ithelial cells repair the kidney after injury.Cell Stem Cell 2: 284291, 2008

    32. Remuzzi G, Cattaneo D, Perico N: The ag-gravating mechanisms of aldosterone onkidney fibrosis. J Am Soc Nephrol 19:14591462, 2008

    33. Leroy V, De Seigneux S, Agassiz V, HaslerU, Rafestin-Oblin ME, Vinciguerra M, Mar-tin PY, Feraille E: Aldosterone activates NF-kappaB in the collecting duct. J Am SocNephrol 20: 131144, 2009

    34. Tu X, Chen X, Xie Y, Shi S, Wang J, Chen Y,Li J: Anti-inflammatory renoprotective ef-fect of clopidogrel and irbesartan inchronic renal injury. J Am Soc Nephrol 19:7783, 2008

    35. Eddy AA: Molecular insights into renal in-terstitial fibrosis. J Am Soc Nephrol 7:24952508, 1996

    36. Haverty TP, Kelly CJ, Hines WH, AmentaPS, Watanabe M, Harper RA, Kefalides NA,Neilson EG: Characterization of a renal tu-bular epithelial cell line which secretes theautologous target antigen of autoimmuneexperimental interstitial nephritis. J CellBiol 107: 13591368, 1988

    37. Howard BV, Macarak EJ, Gunson D,Kefalides NA: Characterization of the col-lagen synthesized by endothelial cells inculture. Proc Natl Acad Sci U S A 73: 23612364, 1976

    38. Eismann U, Sommer M, Kosmehl H, Ap-penroth D, Fleck C, Stein G: Fibronectinsplice variantsprognostic markers for thestage of renal interstitial fibrosis in the rat.Nephron 92: 379388, 2002

    39. Hewitson TD, Darby IA, Bisucci T, JonesCL, Becker GJ:Evolution of tubulointerstitialfibrosis in experimental renal infectionand scar-ring. J Am Soc Nephrol 9: 632642, 1998

    40. Hewitson TD: Renal tubulointerstitial fibro-sis: Common but never simple. Am JPhysiol Renal Physiol 296: F1239F1244,2009

    41. Chen CZ, Raghunath M: Focus on collagen:In vitro systems to study fibrogenesis andantifibrosis state of the art. FibrogenesisTissue Repair 2: 7, 2009

    42. Dallas SL, Chen Q, Sivakumar P: Dynamicsof assembly and reorganization of extracel-lular matrix proteins. Curr Top Dev Biol 75:124, 2006

    43. Kadler KE, Hill A, Canty-Laird EG: Collagenfibrillogenesis: Fibronectin, integrins, andminor collagens as organizers and nuclea-tors. Curr Opin Cell Biol 20: 495501, 2008

    44. Bradshaw AD: The role of SPARC in extra-cellular matrix assembly. J Cell CommunSignal 2009

    45. Holmes DF, Kadler KE: The 104 microfi-bril structure of thin cartilage fibrils. ProcNatl Acad Sci U S A 103: 1724917254,2006

    46. Bradshaw AD, Sage EH: SPARC, a matricel-lular protein that functions in cellular differ-entiation and tissue response to injury.J Clin Invest 107: 10491054, 2001

    47. Tremble PM, Lane TF, Sage EH, Werb Z:SPARC, a secreted protein associated withmorphogenesis and tissue remodeling, in-duces expression of metalloproteinases infibroblasts through a novel extracellularmatrix-dependent pathway. J Cell Biol 121:14331444, 1993

    48. Pichler RH, Hugo C, Shankland SJ, ReedMJ, Bassuk JA, Andoh TF, Lombardi DM,Schwartz SM, Bennett WM, Alpers CE,Sage EH, Johnson RJ, Couser WG: SPARCis expressed in renal interstitial fibrosis andin renal vascular injury. Kidney Int 50:19781989, 1996

    49. Liu Y: New insights into epithelial-mesen-chymal transition in kidney fibrosis. J AmSoc Nephrol 21: 212222, 2010

    50. Zeisberg EM, Potenta SE, Sugimoto H,Zeisberg M, Kalluri R: Fibroblasts in kidneyfibrosis emerge via endothelial-to-mes-enchymal transition. J Am Soc Nephrol19: 22822287, 2008

    51. Zeisberg M, Kalluri R: Fibroblasts emergevia epithelial-mesenchymal transition inchronic kidney fibrosis. Front Biosci 13:69916998, 2008

    52. Neilson EG, McCafferty E, Feldman A,Clayman MD, Zakheim B, Korngold R:Spontaneous interstitial nephritis in kdkdmice. I. An experimental model of autoim-

    mune renal disease. J Immunol 133: 25602565, 1984

    53. Roufosse C, Bou-Gharios G, Prodromidi E,Alexakis C, Jeffery R, Khan S, Otto WR,Alter J, Poulsom R, Cook HT: Bone mar-row-derived cells do not contribute signif-icantly to collagen I synthesis in a murinemodel of renal fibrosis. J Am Soc Nephrol17: 775782, 2006

    54. Humphreys BD, Lin SL, Kobayashi A, Hud-son TE, Nowlin BT, Bonventre JV, ValeriusMT, McMahon AP, Duffield JS: Fate tracingreveals the pericyte and not epithelial ori-gin of myofibroblasts in kidney fibrosis.Am J Pathol 176: 8597, 2010

    55. Lin SL, Kisseleva T, Brenner DA, DuffieldJS: Pericytes and perivascular fibroblastsare the primary source of collagen-produc-ing cells in obstructive fibrosis of the kid-ney. Am J Pathol 173: 16171627, 2008

    56. Niedermeier M, Reich B, Rodriguez GomezM, Denzel A, Schmidbauer K, Gobel N,Talke Y, Schweda F, Mack M: CD4 T cellscontrol the differentiation of Gr1 mono-cytes into fibrocytes. Proc Natl Acad SciU S A 106: 1789217897, 2009

    57. Taguchi T, Razzaque MS: The collagen-spe-cific molecular chaperone HSP47: Is there arole in fibrosis? Trends Mol Med 13: 4553,2007

    58. Okada H, Ban S, Nagao S, Takahashi H,Suzuki H, Neilson EG: Progressive renal fi-brosis in murine polycystic kidney disease:An immunohistochemical observation. Kid-ney Int 58: 587597, 2000

    59. Iwano M, Plieth D, Danoff TM, Xue C,Okada H, Neilson EG: Evidence that fibro-blasts derive from epithelium during tissuefibrosis. J Clin Invest 110: 341350, 2002

    60. Inoue T, Takenaka T, Hayashi M, MonkawaT, Yoshino J, Shimoda K, Neilson EG, Su-zuki H, Okada H: Fibroblast expression ofan IB-dominant negative transgene atten-uates renal fibrosis. J Am Soc Nephrol2010, in press

    61. Iwano M, Fischer A, Okada H, Plieth D, XueC, Danoff TM, Neilson EG: Conditionalabatement of tissue fibrosis using nucleo-side analogs to selectively corrupt DNAreplication in transgenic fibroblasts. MolTher 3: 149159, 2001

    62. McDougall S, Dallon J, Sherratt J, Maini P:Fibroblast migration and collagen deposi-tion during dermal wound healing: Mathe-matical modelling and clinical implications.Philos Transact A Math Phys Eng Sci 364:13851405, 2006

    63. Whitby DJ, Ferguson MW: Immunohisto-chemical localization of growth factors infetal wound healing. Dev Biol 147: 207215, 1991

    64. Schnaper HW, Jandeska S, Runyan CE,Hubchak SC, Basu RK, Curley JF, Smith RD,Hayashida T: TGF-beta signal transductionin chronic kidney disease. Front Biosci 14:24482465, 2009

    BRIEF REVIEWwww.jasn.org

    J Am Soc Nephrol 21: 18191834, 2010 Mechanisms of Tubulointerstitial Fibrosis 1827

  • 65. Murphy M, Docherty NG, Griffin B, HowlinJ, McArdle E, McMahon R, Schmid H, Kret-zler M, Droguett A, Mezzano S, Brady HR,Furlong F, Godson C, Martin F: IHG-1 am-plifies TGF-beta1 signaling and is in-creased in renal fibrosis. J Am Soc Nephrol19: 16721680, 2008

    66. Kato M, Arce L, Natarajan R: MicroRNAsand their role in progressive kidney dis-eases. Clin J Am Soc Nephrol 4: 12551266, 2009

    67. Noh H, Oh EY, Seo JY, Yu MR, Kim YO, HaH, Lee HB: Histone deacetylase-2 is a keyregulator of diabetes- and transforminggrowth factor-beta1-induced renal injury.Am J Physiol Renal Physiol 297: F729F739, 2009

    68. Bechtel W, McGoohan S, Zeisberg EM,Muller GA, Kalbacher H, Salant DJ, MullerCA, Kalluri R, Zeisberg M: Methylation de-termines fibroblast activation and fibrogen-esis in the kidney. Nat Med 16: 544550,2010

    69. Rosenbloom J, Castro SV, Jimenez SA:Narrative review: Fibrotic diseases: Cellularand molecular mechanisms and novel ther-apies. Ann Intern Med 152: 159166, 2010

    70. Wynn TA: Cellular and molecular mecha-nisms of fibrosis. J Pathol 214: 199210,2008

    71. Border WA, Noble NA: Interactions oftransforming growth factor-beta and an-giotensin II in renal fibrosis. Hypertension31: 181188, 1998

    72. Matsuo S, Lopez-Guisa JM, Cai X, Oka-mura DM, Alpers CE, Bumgarner RE, Pe-ters MA, Zhang G, Eddy AA: Multifunction-ality of PAI-1 in fibrogenesis: Evidencefrom obstructive nephropathy in PAI-1-overexpressing mice. Kidney Int 67: 22212238, 2005

    73. Ruster C, Wolf G: Renin-angiotensin-aldo-sterone system and progression of renaldisease. J Am Soc Nephrol 17: 29852991,2006

    74. Border WA, Okuda S, Languino LR, SpornMB, Ruoslahti E: Suppression of experi-mental glomerulonephritis by antiserumagainst transforming growth factor beta 1.Nature 346: 371374, 1990

    75. Sharma K, Jin Y, Guo J, Ziyadeh FN: Neu-tralization of TGF-beta by anti-TGF-betaantibody attenuates kidney hypertrophyand the enhanced extracellular matrix geneexpression in STZ-induced diabetic mice.Diabetes 45: 522530, 1996

    76. Cosgrove D, Rodgers K, Meehan D, MillerC, Bovard K, Gilroy A, Gardner H, Kotelian-ski V, Gotwals P, Amatucci A, Kalluri R:Integrin alpha1beta1 and transforminggrowth factor-beta1 play distinct roles inalport glomerular pathogenesis and serveas dual targets for metabolic therapy. Am JPathol 157: 16491659, 2000

    77. Isaka Y, Brees DK, Ikegaya K, Kaneda Y,Imai E, Noble NA, Border WA: Gene ther-

    apy by skeletal muscle expression ofdecorin prevents fibrotic disease in rat kid-ney. Nat Med 2: 418423, 1996

    78. Ziyadeh FN, Hoffman BB, Han DC, Iglesias-DeLa Cruz MC, Hong SW, Isono M, Chen S,McGowan TA, Sharma K: Long-term preven-tion of renal insufficiency, excess matrix geneexpression, and glomerular mesangial matrixexpansion by treatment with monoclonal anti-transforming growth factor-beta antibody indb/db diabetic mice. Proc Natl Acad Sci U S A97: 80158020, 2000

    79. Hugo C: The thrombospondin 1-TGF-betaaxis in fibrotic renal disease. Nephrol DialTransplant 18: 12411245, 2003

    80. Zeisberg M, Hanai J, Sugimoto H, Mam-moto T, Charytan D, Strutz F, Kalluri R:BMP-7 counteracts TGF-beta1-induced ep-ithelial-to-mesenchymal transition and re-verses chronic renal injury. Nat Med 9:964968, 2003

    81. Gewin L, Bulus N, Mernaugh G, MoeckelG, Harris RC, Moses HL, Pozzi A, Zent R:TGF-{beta} receptor deletion in the renalcollecting system exacerbates fibrosis.J Am Soc Nephrol 21: 13341343, 2010

    82. Kopp JB: TGF signaling and the renaltubular epithelial cell: Too much, too little,and just right. J Am Soc Nephrol 21: 12411243, 2010

    83. Mott JD, Werb Z: Regulation of matrix bi-ology by matrix metalloproteinases. CurrOpin Cell Biol 16: 558564, 2004

    84. Cheng S, Lovett DH: Gelatinase A (MMP-2)is necessary and sufficient for renal tubularcell epithelial-mesenchymal transforma-tion. Am J Pathol 162: 19371949, 2003

    85. Sabeh F, Li XY, Saunders TL, Rowe RG,Weiss SJ: Secreted versus membrane-an-chored collagenases: Relative roles in fi-broblast-dependent collagenolysis and in-vasion. J Biol Chem 284: 2300123011,2009

    86. Catania JM, Chen G, Parrish AR: Role ofmatrix metalloproteinases in renal patho-physiologies. Am J Physiol Renal Physiol292: F905F911, 2007

    87. Ronco P, Chatziantoniou C: Matrix metal-loproteinases and matrix receptors inprogression and reversal of kidney dis-ease: therapeutic perspectives. KidneyInt 74: 873878, 2008

    88. Zeisberg M, Khurana M, Rao VH, CosgroveD, Rougier JP, Werner MC, Shield CF,Werb Z, Kalluri R: Stage-specific action ofmatrix metalloproteinases influences pro-gressive hereditary kidney disease. PLoSMed 3: e100, 2006

    89. Cheng S, Pollock AS, Mahimkar R, OlsonJL, Lovett DH: Matrix metalloproteinase 2and basement membrane integrity: A uni-fying mechanism for progressive renal in-jury. FASEB J 20: 18981900, 2006

    90. Kassiri Z, Oudit GY, Kandalam V, Awad A,Wang X, Ziou X, Maeda N, HerzenbergAM, Scholey JW: Loss of TIMP3 enhances

    interstitial nephritis and fibrosis. J Am SocNephrol 20: 12231235, 2009

    91. Ota I, Li XY, Hu Y, Weiss SJ: Induction ofa MT1-MMP and MT2-MMP-dependentbasement membrane transmigration pro-gram in cancer cells by Snail1. Proc NatlAcad Sci U S A 106: 20318 20323,2009

    92. Rowe RG, Li XY, Hu Y, Saunders TL, Vir-tanen I, Garcia de Herreros A, Becker KF,Ingvarsen S, Engelholm LH, Bommer GT,Fearon ER, Weiss SJ: Mesenchymal cellsreactivate Snail1 expression to drive three-dimensional invasion programs. J Cell Biol184: 399408, 2009

    93. Eddy AA, Fogo AB: Plasminogen activatorinhibitor-1 in chronic kidney disease: Evi-dence and mechanisms of action. J Am SocNephrol 17: 29993012, 2006

    94. Eddy AA: Plasminogen activator inhibitor-1and the kidney. Am J Physiol Renal Physiol283: F209F220, 2002

    95. Birkedal-Hansen H, Taylor RE, Bhown AS,Katz J, Lin HY, Wells BR: Cleavage of bo-vine skin type III collagen by proteolyticenzymes. Relative resistance of the fibrillarform. J Biol Chem 260: 1641116417, 1985

    96. Nagase H: Activation mechanisms of ma-trix metalloproteinases. Biol Chem 378:151160, 1997

    97. Zhang G, Kernan KA, Collins SJ, Cai X,Lopez-Guisa JM, Degen JL, Shvil Y, EddyAA: Plasmin(ogen) promotes renal inter-stitial fibrosis by promoting epithelial-to-mesenchymal transition: role of plasmin-activated signals. J Am Soc Nephrol 18:846859, 2007

    98. Yang J, Shultz RW, Mars WM, Wegner RE,Li Y, Dai C, Nejak K, Liu Y: Disruption oftissue-type plasminogen activator gene inmice reduces renal interstitial fibrosis in ob-structive nephropathy. J Clin Invest 110:15251538, 2002

    99. Zhang G, Kim H, Cai X, Lopez-Guisa JM,Alpers CE, Liu Y, Carmeliet P, Eddy AA:Urokinase receptor deficiency acceleratesrenal fibrosis in obstructive nephropathy.J Am Soc Nephrol 14: 12541271, 2003

    100. Fintha A, Sebe A, Masszi A, Terebessy T,Huszar T, Rosivall L, Mucsi I: Angiotensin IIactivates plasminogen activator inhibitor-Ipromoter in renal tubular epithelial cells viathe AT1 receptor. Acta Physiol Hung 94:1930, 2007

    101. Jung GS, Kim MK, Choe MS, Lee KM, KimHS, Park YJ, Choi HS, Lee KU, Park KG, LeeIK: The orphan nuclear receptor SHP atten-uates renal fibrosis. J Am Soc Nephrol 20:21622170, 2009

    102. Oda T, Jung YO, Kim HS, Cai X, Lopez-Guisa JM, Ikeda Y, Eddy AA: PAI-1 defi-ciency attenuates the fibrogenic responseto ureteral obstruction. Kidney Int 60: 587596, 2001

    103. Krag S, Danielsen CC, Carmeliet P, Nyen-gaard J, Wogensen L: Plasminogen activa-

    BRIEF REVIEW www.jasn.org

    1828 Journal of the American Society of Nephrology J Am Soc Nephrol 21: 18191834, 2010

  • tor inhibitor-1 gene deficiency attenuatesTGF-beta1-induced kidney disease. KidneyInt 68: 26512666, 2005

    104. Zeisberg M, Bottiglio C, Kumar N, Mae-shima Y, Strutz F, Muller GA, Kalluri R:Bone morphogenic protein-7 inhibits pro-gression of chronic renal fibrosis associatedwith two genetic mouse models. Am JPhysiol Renal Physiol 285: F1060F1067,2003

    105. Sugimoto H, Grahovac G, Zeisberg M, Kal-luri R: Renal fibrosis and glomerulosclerosisin a new mouse model of diabetic ne-phropathy and its regression by bone mor-phogenic protein-7 and advanced glyca-tion end product inhibitors. Diabetes 56:18251833, 2007

    106. Adamczak M, Gross ML, Krtil J, Koch A,Tyralla K, Amann K, Ritz E: Reversal of glo-merulosclerosis after high-dose enalapriltreatment in subtotally nephrectomizedrats. J Am Soc Nephrol 14: 28332842,2003

    107. Thornhill BA, Forbes MS, Marcinko ES,Chevalier RL: Glomerulotubular disconnec-tion in neonatal mice after relief of partialureteral obstruction. Kidney Int 72: 11031112, 2007

    108. Everts V, van der Zee E, Creemers L, Beer-tsen W: Phagocytosis and intracellular di-gestion of collagen, its role in turnover andremodelling. Histochem J 28: 229245,1996

    109. Hilliard LM, Russo LM, Comper WD: Hyper-tension-mediated albuminuria is associ-ated with reduced lysosomal activity in thekidney and the heart. Am J Nephrol 29:454464, 2009

    110. Shimoda N, Fukazawa N, Nonomura K,Fairchild RL: Cathepsin g is required forsustained inflammation and tissue injury af-ter reperfusion of ischemic kidneys. Am JPathol 170: 930940, 2007

    111. Sebekova K, Paczek L, Dammrich J, Ling H,Spustova V, Gaciong Z, Heidland A: Effectsof protease therapy in the remnant kidneymodel of progressive renal failure. MinerElectrolyte Metab 23: 291295, 1997

    112. Huang S, Schaefer RM, Reisch S, Paczek L,Schaefer L, Teschner M, Sebekova K, Hei-dland A: Suppressed activities of cathep-sins and metalloproteinases in the chronicmodel of puromycin aminonucleoside ne-phrosis. Kidney Blood Press Res 22: 121127, 1999

    113. Vielhauer V, Berning E, Eis V, Kretzler M,Segerer S, Strutz F, Horuk R, Grone HJ,Schlondorff D, Anders HJ: CCR1 blockadereduces interstitial inflammation and fi-brosis in mice with glomerulosclerosisand nephrotic syndrome. Kidney Int 66:22642278, 2004

    114. John R, Nelson PJ: Dendritic cells in thekidney. J Am Soc Nephrol 18: 26282635,2007

    115. Kelly CJ: Cellular immunity and the tubu-

    lointerstitium. Semin Nephrol 19: 182187,1999

    116. Lebleu VS, Sugimoto H, Miller CA, GattoneVH, 2nd, Kalluri R: Lymphocytes are dis-pensable for glomerulonephritis but re-quired for renal interstitial fibrosis in matrixdefect-induced Alport renal disease. LabInvest 88: 284292, 2008

    117. Sean Eardley K, Cockwell P: Macrophagesand progressive tubulointerstitial disease.Kidney Int 68: 437455, 2005

    118. Holdsworth SR, Summers SA: Role of mastcells in progressive renal diseases. J AmSoc Nephrol 19: 22542261, 2008

    119. Pulskens WP, Rampanelli E, Teske GJ, But-ter LM, Claessen N, Luirink IK, Poll TV,Florquin S, Leemans, JC: TLR4 promotesfibrosis but attenuates tubular damage inprogressive renal injury. J Am Soc Nephrol21: 12991308, 2010

    120. Anders HJ, Vielhauer V, Eis V, Linde Y,Kretzler M, Perez de Lema G, Strutz F,Bauer S, Rutz M, Wagner H, Grone HJ,Schlondorff D: Activation of toll-like recep-tor-9 induces progression of renal diseasein MRL-Fas(lpr) mice. FASEB J 18: 534536, 2004

    121. Leemans JC, Butter LM, Pulskens WP,Teske GJ, Claessen N, van der Poll T,Florquin S: The role of Toll-like receptor 2in inflammation and fibrosis during pro-gressive renal injury. PLoS One 4: e5704,2009

    122. Tapmeier TT, Fearn A, Brown K,Chowdhury P, Sacks SH, Sheerin NS, WongW: Pivotal role of CD4() T cells in renalfibrosis following ureteric obstruction. Kid-ney Int 78: 351362, 2010

    123. Kelly CJ, Zurier RB, Krakauer KA, BlanchardN, Neilson EG: Prostaglandin E1 inhibitseffector T cell induction and tissue damagein experimental murine interstitial nephritis.J Clin Invest 79: 782789, 1987

    124. Ascon M, Ascon DB, Liu M, Cheadle C,Sarkar C, Racusen L, Hassoun HT, Rabb H:Renal ischemia-reperfusion leads to longterm infiltration of activated and effector-memory T lymphocytes. Kidney Int 75:526535, 2009

    125. Burne-Taney MJ, Liu M, Ascon D, Molls RR,Racusen L, Rabb H: Transfer of lympho-cytes from mice with renal ischemia caninduce albuminuria in naive mice: A possi-ble mechanism linking early injury and pro-gressive renal disease? Am J Physiol RenalPhysiol 291: F981F986, 2006

    126. Gandolfo MT, Jang HR, Bagnasco SM, KoGJ, Agreda P, Satpute SR, Crow MT, KingLS, Rabb H: Foxp3 regulatory T cells par-ticipate in repair of ischemic acute kidneyinjury. Kidney Int 76: 717729, 2009

    127. Heller F, Lindenmeyer MT, Cohen CD,Brandt U, Draganovici D, Fischereder M,Kretzler M, Anders HJ, Sitter T, MosbergerI, Kerjaschki D, Regele H, Schlondorff D,Segerer S: The contribution of B cells to

    renal interstitial inflammation. Am J Pathol170: 457468, 2007

    128. Segerer S, Schlondorff D: B cells and ter-tiary lymphoid organs in renal inflamma-tion. Kidney Int 73: 533537, 2008

    129. Ricardo SD, van Goor H, Eddy AA: Macro-phage diversity in renal injury and repair.J Clin Invest 118: 35223530, 2008

    130. Nishida M, Hamaoka K: Macrophage phe-notype and renal fibrosis in obstructive ne-phropathy. Nephron Exp Nephrol 110:e31e36, 2008

    131. Ko GJ, Boo CS, Jo SK, Cho WY, Kim HK:Macrophages contribute to the develop-ment of renal fibrosis following ischaemia/reperfusion-induced acute kidney injury.Nephrol Dial Transplant 23: 842852, 2008

    132. Duffield JS, Forbes SJ, Constandinou CM,Clay S, Partolina M, Vuthoori S, Wu S, LangR, Iredale JP: Selective depletion of mac-rophages reveals distinct, opposing rolesduring liver injury and repair. J Clin Invest115: 5665, 2005

    133. Wang Y, Wang YP, Zheng G, Lee VW, Ouy-ang L, Chang DH, Mahajan D, Coombs J,Wang YM, Alexander SI, Harris DC: Ex vivoprogrammed macrophages ameliorate ex-perimental chronic inflammatory renal dis-ease. Kidney Int 72: 290299, 2007

    134. Macconi D, Chiabrando C, Schiarea S, Ai-ello S, Cassis L, Gagliardini E, Noris M,Buelli S, Zoja C, Corna D, Mele C, Fanelli R,Remuzzi G, Benigni A: Proteasomal pro-cessing of albumin by renal dendritic cellsgenerates antigenic peptides. J Am SocNephrol 20: 123130, 2009

    135. Miyazawa S, Hotta O, Doi N, Natori Y,Nishikawa K, Natori Y: Role of mast cells inthe development of renal fibrosis: Use ofmast cell-deficient rats. Kidney Int 65:22282237, 2004

    136. Anders HJ, Belemezova E, Eis V, SegererS, Vielhauer V, Perez de Lema G, KretzlerM, Cohen CD, Frink M, Horuk R, HudkinsKL, Alpers CE, Mampaso F, SchlondorffD: Late onset of treatment with a chemo-kine receptor CCR1 antagonist preventsprogression of lupus nephritis in MRL-Fas(lpr) mice. J Am Soc Nephrol 15:15041513, 2004

    137. Jones LK,OSullivan KM, Semple T, KuligowskiMP, Fukami K, Ma FY, Nikolic-Paterson DJ,Holdsworth SR, Kitching AR: IL-1RI deficiencyameliorates early experimental renal interstitialfibrosis. Nephrol Dial Transplant 24:30243032, 2009

    138. Khan SB, Cook HT, Bhangal G, Smith J,Tam FW, Pusey CD: Antibody blockade ofTNF-alpha reduces inflammation and scar-ring in experimental crescentic glomerulo-nephritis. Kidney Int 67: 18121820, 2005

    139. Muller GA, Rodemann HP: Characteriza-tion of human renal fibroblasts in healthand disease: I. Immunophenotyping of cul-tured tubular epithelial cells and fibroblastsderived from kidneys with histologically

    BRIEF REVIEWwww.jasn.org

    J Am Soc Nephrol 21: 18191834, 2010 Mechanisms of Tubulointerstitial Fibrosis 1829

  • proven interstitial fibrosis. Am J Kidney Dis17: 680683, 1991

    140. Rodemann HP, Muller GA: Characteriza-tion of human renal fibroblasts in healthand disease: II. In vitro growth, differentia-tion, and collagen synthesis of fibroblastsfrom kidneys with interstitial fibrosis. Am JKidney Dis 17: 684686, 1991

    141. Kaissling B, Le Hir M: The renal corticalinterstitium: morphological and functionalaspects. Histochem Cell Biol 130: 247262,2008

    142. Grande MT, Lopez-Novoa JM: Fibroblastactivation and myofibroblast generation inobstructive nephropathy. Nat Rev Nephrol5: 319328, 2009

    143. Kalluri R, Zeisberg M: Fibroblasts in cancer.Nat Rev Cancer 6: 392401, 2006

    144. Bayreuther K, Rodemann HP, Hommel R,Dittmann K, Albiez M, Francz PI: Humanskin fibroblasts in vitro differentiate along aterminal cell lineage. Proc Natl Acad Sci US A 85: 51125116, 1988

    145. Schor SL, Schor AM: Clonal heterogeneityin fibroblast phenotype: Implications forthe control of epithelial-mesenchymal in-teractions. Bioessays 7: 200204, 1987

    146. Kalluri R, Neilson EG: Epithelial-mesenchy-mal transition and its implications for fibro-sis. J Clin Invest 112: 17761784, 2003

    147. Zeisberg M, Neilson EG: Biomarkers forepithelial-mesenchymal transitions. J ClinInvest 119: 14291437, 2009

    148. Hutchison N, Hendry BM, Sharpe CC: Rhoisoforms have distinct and specific func-tions in the process of epithelial to mesen-chymal transition in renal proximal tubularcells. Cell Signal 21: 15221531, 2009

    149. Morikawa S, Baluk P, Kaidoh T, Haskell A,Jain RK, McDonald DM: Abnormalities inpericytes on blood vessels and endothelialsprouts in tumors. Am J Pathol 160: 9851000, 2002

    150. Diaz-Flores L, Gutierrez R, Madrid JF,Varela H, Valladares F, Acosta E, Martin-Vasallo P, Diaz-Flores, L Jr.: Pericytes. Mor-phofunction, interactions and pathology ina quiescent and activated mesenchymalcell niche. Histol Histopathol 24: 909969,2009

    151. Neilson EG: Mechanisms of disease: Fibro-blastsa new look at an old problem. NatClin Pract Nephrol 2: 101108, 2006

    152. Okada H, Inoue T, Kanno Y, Kobayashi T,Ban S, Kalluri R, Suzuki H: Renal fibroblast-like cells in Goodpasture syndrome rats.Kidney Int 60: 597606, 2001

    153. Zeisberg M, Strutz F, Muller GA: Role offibroblast activation in inducing interstitialfibrosis. J Nephrol 13 [Suppl 3]: S111S120, 2000

    154. Gabbiani G: The myofibroblast in woundhealing and fibrocontractive diseases.J Pathol 200: 500503, 2003

    155. Hay ED: The mesenchymal cell, its role inthe embryo, and the remarkable signaling

    mechanisms that create it. Dev Dyn 233:706720, 2005

    156. Takeji M, Moriyama T, Oseto S, Kawada N,Hori M, Imai E, Miwa T: Smooth musclealpha-actin deficiency in myofibroblastsleads to enhanced renal tissue fibrosis.J Biol Chem 281: 4019340200, 2006

    157. Strutz F, Okada H, Lo CW, Danoff T, Ca-rone RL, Tomaszewski JE, Neilson EG:Identification and characterization of a fi-broblast marker: FSP1. J Cell Biol 130:393405, 1995

    158. Le Hir M, Hegyi I, Cueni-Loffing D, Loff-ing J, Kaissling B: Characterization of re-nal interstitial fibroblast-specific protein1/S100A4-positive cells in healthy and in-flamed rodent kidneys. Histochem CellBiol 123: 335346, 2005

    159. Li L, Zepeda-Orozco D, Black R, Lin F: Au-tophagy is a component of epithelial cellfate in obstructive uropathy. Am J Pathol176: 17671778, 2010

    160. Zeisberg M, Duffield JS: Resolved: EMTproduces fibroblasts in the kidney. J AmSoc Nephrol 21: 12471253, 2010

    161. Inoue T, Plieth D, Venkov CD, Xu C, Neil-son EG: Antibodies against macrophagesthat overlap in specificity with fibroblasts.Kidney Int 67: 24882493, 2005

    162. Trimboli AJ, Cantemir-Stone CZ, Li F,Wallace JA, Merchant A, Creasap N,Thompson JC, Caserta E, Wang H,Chong JL, Naidu S, Wei G, Sharma SM,Stephens JA, Fernandez SA, Gurcan MN,Weinstein MB, Barsky SH, Yee L, RosolTJ, Stromberg PC, Robinson ML, Pepin F,Hallett M, Park M, Ostrowski MC, LeoneG: Pten in stromal fibroblasts suppressesmammary epithelial tumours. Nature 461:10841091, 2009

    163. Floege J, Eitner F, Alpers CE: A new look atplatelet-derived growth factor in renal dis-ease. J Am Soc Nephrol 19: 1223, 2008

    164. Krupa A, Jenkins R, Luo DD, Lewis A, Phil-lips A, Fraser D: Loss of microRNA-192 pro-motes fibrogenesis in diabetic nephropa-thy. J Am Soc Nephrol 21: 438447, 2010

    165. Higgins DF, Kimura K, Bernhardt WM,Shrimanker N, Akai Y, Hohenstein B,Saito Y, Johnson RS, Kretzler M, CohenCD, Eckardt KU, Iwano M, Haase VH:Hypoxia promotes fibrogenesis in vivovia HIF-1 stimulation of epithelial-to-mesenchymal transition. J Clin Invest117: 38103820, 2007

    166. Zeisberg EM, Tarnavski O, Zeisberg M, Dorf-man AL, McMullen JR, Gustafsson E, Chan-draker A, Yuan X, Pu WT, Roberts AB, NeilsonEG, Sayegh MH, Izumo S, Kalluri R: Endotheli-al-to-mesenchymal transition contributes tocardiac fibrosis. Nat Med 13: 952961, 2007

    167. Wilm B, Ipenberg A, Hastie ND, Burch JB,Bader DM: The serosal mesothelium is amajor source of smooth muscle cells of thegut vasculature. Development 132: 53175328, 2005

    168. Que J, Wilm B, Hasegawa H, Wang F,Bader D, Hogan BL: Mesothelium contrib-utes to vascular smooth muscle and mes-enchyme during lung development. ProcNatl Acad Sci U S A 105: 1662616630,2008

    169. Eitner F, Bucher E, van Roeyen C, Kunter U,Rong S, Seikrit C, Villa L, Boor P, Fredriks-son L, Backstrom G, Eriksson U, Ostman A,Floege J, Ostendorf T: PDGF-C is a proin-flammatory cytokine that mediates renal in-terstitial fibrosis. J Am Soc Nephrol 19:281289, 2008

    170. Boor P, Konieczny A, Villa L, Kunter U, vanRoeyen CR, LaRochelle WJ, Smithson G,Arrol S, Ostendorf T, Floege J: PDGF-Dinhibition by CR002 ameliorates tubulo-interstitial fibrosis following experimentalglomerulonephritis. Nephrol Dial Trans-plant 22: 13231331, 2007

    171. Xin J, Homma T, Matsusaka T, Ma J, Isaka Y,Imai E, Ichikawa I: Suppression of cyclospor-ine a nephrotoxicity in vivo by transforminggrowth factor beta receptor-immunoglobulinG chimeric protein. Transplantation 77:14331442, 2004

    172. Benigni A, Zoja C, Corna D, Zatelli C, ContiS, Campana M, Gagliardini E, Rottoli D,Zanchi C, Abbate M, Ledbetter S, RemuzziG: Add-on anti-TGF-beta antibody to ACEinhibitor arrests progressive diabetic ne-phropathy in the rat. J Am Soc Nephrol 14:18161824, 2003

    173. Miric G, Dallemagne C, Endre Z, MargolinS, Taylor SM, Brown L: Reversal of cardiacand renal fibrosis by pirfenidone and spi-ronolactone in streptozotocin-diabetic rats.Br J Pharmacol 133: 687694, 2001

    174. RamachandraRao SP, Zhu Y, Ravasi T,McGowan TA, Toh I, Dunn SR, Okada S,Shaw MA, Sharma K: Pirfenidone is reno-protective in diabetic kidney disease. J AmSoc Nephrol 20: 17651775, 2009

    175. Shimizu T, Fukagawa M, Kuroda T, Hata S,Iwasaki Y, Nemoto M, Shirai K, Yamauchi S,Margolin SB, Shimizu F, Kurokawa K: Pir-fenidone prevents collagen accumulation inthe remnant kidney in rats with partial nephrec-tomy. Kidney Int Suppl 63: S239S243, 1997

    176. Waanders F, van den Berg E, Nagai R, vanVeen I, Navis G, van Goor H: Renoprotec-tive effects of the AGE-inhibitor pyridox-amine in experimental chronic allograft ne-phropathy in rats. Nephrol Dial Transplant23: 518524, 2008

    177. Moon JA, Kim HT, Cho IS, Sheen YY, KimDK: IN-1130, a novel transforming growthfactor-beta type I receptor kinase (ALK5)inhibitor, suppresses renal fibrosis in ob-structive nephropathy. Kidney Int 70:12341243, 2006

    178. Tan X, Li Y, Liu Y: Paricalcitol attenuatesrenal interstitial fibrosis in obstructive ne-phropathy. J Am Soc Nephrol 17: 33823393, 2006

    179. Sharpe CC, Dockrell ME, Scott R, Noor MI,

    BRIEF REVIEW www.jasn.org

    1830 Journal of the American Society of Nephrology J Am Soc Nephrol 21: 18191834, 2010

  • Cowsert LM, Monia BP, Hendry BM: Evi-dence of a role for Ki-Ras in the stimulatedproliferation of renal fibroblasts. J Am SocNephrol 10: 11861192, 1999

    180. Okada H, Watanabe Y, Inoue T, KobayashiT, Kikuta T, Kanno Y, Ban S, Suzuki H: An-giotensin II type 1 receptor blockade atten-uates renal fibrogenesis in an immune-mediated nephritic kidney throughcounter-activation of angiotensin II type 2receptor. Biochem Biophys Res Commun314: 403408, 2004

    181. Yang L, Besschetnova TY, Brooks CR, ShahJV, Bonventre JV: Epithelial cell cycle arrestin G2/M mediates kidney fibrosis after in-jury. Nat Med 16: 535543, 2010

    182. Zeisberg M, Strutz F, Muller GA: Renal fi-brosis: An update. Curr Opin Nephrol Hy-pertens 10: 315320, 2001

    183. Wang S, LaPage J, Hirschberg R: Protein-uria and progression of chronic renal dis-ease. Kidney Blood Press Res 23: 167169,2000

    184. Baines RJ, Brunskill NJ: The molecular in-teractions between filtered proteins andproximal tubular cells in proteinuria.Nephron Exp Nephrol 110: e67e71, 2008

    185. Schlondorff DO: Overview of factors con-tributing to the pathophysiology of pro-gressive renal disease. Kidney Int 74: 860866, 2008

    186. Pichler R, Giachelli C, Young B, Alpers CE,Couser WG, Johnson RJ: The pathogenesisof tubulointerstitial disease associated withglomerulonephritis: The glomerular cyto-kine theory. Miner Electrolyte Metab 21:317327, 1995

    187. Hirschberg R, Wang S: Proteinuria andgrowth factors in the development of tubu-lointerstitial injury and scarring in kidneydisease. Curr Opin Nephrol Hypertens 14:4352, 2005

    188. Rosivall L, Mirzahosseini S, Toma I, Sipos A,Peti-Peterdi J: Fluid flow in the juxtaglo-merular interstitium visualized in vivo. Am JPhysiol Renal Physiol 291: F1241F1247,2006

    189. Rohatgi R, Flores D: Intratubular hydrody-namic forces influence tubulointerstitial fi-brosis in the kidney. Curr Opin NephrolHypertens 19: 6571, 2010

    190. Sanz AB, Sanchez-Nino MD, Ramos AM,Moreno JA, Santamaria B, Ruiz-Ortega M,Egido J, Ortiz A: NF-{kappa}B in renal in-flammation. J Am Soc Nephrol 21: 12541262, 2010

    191. Abbate M, Zoja C, Remuzzi G: How doesproteinuria cause progressive renal dam-age? J Am Soc Nephrol 17: 29742984,2006

    192. Sanchez-Lopez E, Rayego S, Rodrigues-Diez R, Rodriguez JS, Rodriguez-Vita J,Carvajal G, Aroeira LS, Selgas R, MezzanoSA, Ortiz A, Egido J, Ruiz-Ortega M: CTGFpromotes inflammatory cell infiltration ofthe renal interstitium by activating NF-kap-

    paB. J Am Soc Nephrol 20: 15131526,2009

    193. Pontrelli P, Ursi M, Ranieri E, CapobiancoC, Schena FP, Gesualdo L, Grandaliano G:CD40L proinflammatory and profibrotic ef-fects on proximal tubular epithelial cells:Role of NF-kappaB and lyn. J Am SocNephrol 17: 627636, 2006

    194. Li XC, Zhuo JL: Nuclear factor-kappaB as ahormonal intracellular signaling molecule:focus on angiotensin II-induced cardiovas-cular and renal injury. Curr Opin NephrolHypertens 17: 3743, 2008

    195. Esteban V, Lorenzo O, Ruperez M, SuzukiY, Mezzano S, Blanco J, Kretzler M, SugayaT, Egido J, Ruiz-Ortega M: Angiotensin II,via AT1 and AT2 receptors and NF-kappaBpathway, regulates the inflammatory re-sponse in unilateral ureteral obstruction.J Am Soc Nephrol 15: 15141529, 2004

    196. Zoja C, Garcia PB, Remuzzi G: The role ofchemokines in progressive renal disease.Front Biosci 14: 18151822, 2009

    197. Semedo P, Correa-Costa M, Antonio Cene-deze M, Maria Avancini Costa Malheiros D,Antonia dos Reis M, Shimizu MH, SeguroAC, Pacheco-Silva A, Saraiva Camara NO:Mesenchymal stem cells attenuate renal fi-brosis through immune modulation and re-modeling properties in a rat remnant kid-ney model. Stem Cells 27: 30633073,2009

    198. Shimizu H, Maruyama S, Yuzawa Y, Kato T,Miki Y, Suzuki S, Sato W, Morita Y, Ma-ruyama H, Egashira K, Matsuo S: Anti-monocyte chemoattractant protein-1 genetherapy attenuates renal injury induced byprotein-overload proteinuria. J Am SocNephrol 14: 14961505, 2003

    199. Tashiro K, Tamada S, Kuwabara N,Komiya T, Takekida K, Asai T, Iwao H,Sugimura K, Matsumura Y, Takaoka M,Nakatani T, Miura K: Attenuation of renalfibrosis by proteasome inhibition in ratobstructive nephropathy: Possible role ofnuclear factor kappaB. Int J Mol Med 12:587592, 2003

    200. Gong R, Rifai A, Tolbert EM, Biswas P,Centracchio JN, Dworkin LD: Hepatocytegrowth factor ameliorates renal intersti-tial inflammation in rat remnant kidney bymodulating tubular expression of macro-phage chemoattractant protein-1 andRANTES. J Am Soc Nephrol 15: 28682881, 2004

    201. Ka SM, Huang XR, Lan HY, Tsai PY, YangSM, Shui HA, Chen A: Smad7 gene therapyameliorates an autoimmune crescentic glo-merulonephritis in mice. J Am Soc Nephrol18: 17771788, 2007

    202. Lan HY: Smad7 as a therapeutic agent forchronic kidney diseases. Front Biosci 13:49844992, 2008

    203. Tan X, Wen X, Liu Y: Paricalcitol inhibitsrenal inflammation by promoting vitamin Dreceptor-mediated sequestration of NF-

    kappaB signaling. J Am Soc Nephrol 19:17411752, 2008

    204. Takase O, Hirahashi J, Takayanagi A,Chikaraishi A, Marumo T, Ozawa Y, HayashiM, Shimizu N, Saruta T: Gene transfer oftruncated IkappaBalpha prevents tubuloin-terstitial injury. Kidney Int 63: 501513,2003

    205. Herrero-Fresneda I, Torras J, Franquesa M,Vidal A, Cruzado JM, Lloberas N, Fillat C,Grinyo JM: HGF gene therapy attenuatesrenal allograft scarring by preventing theprofibrotic inflammatory-induced mecha-nisms. Kidney Int 70: 265274, 2006

    206. Han SY, Kim CH, Kim HS, Jee YH, Song HK,Lee MH, Han KH, Kim HK, Kang YS, HanJY, Kim YS, Cha DR: Spironolactone pre-vents diabetic nephropathy through ananti-inflammatory mechanism in type 2 di-abetic rats. J Am Soc Nephrol 17: 13621372, 2006

    207. Kim KH, Lee ES, Cha SH, Park JH, Park JS,Chang YC, Park KK: Transcriptional regula-tion of NF-kappaB by ring type decoy oli-godeoxynucleotide in an animal model ofnephropathy. Exp Mol Pathol 86: 114120,2009

    208. Peng M, Jarett L, Meade R, Madaio MP,Hancock WW, George AL Jr., Neilson EG,Gasser DL: Mutant prenyltransferase-likemitochondrial protein (PLMP) and mito-chondrial abnormalities in kd/kd mice. Kid-ney Int 66: 2028, 2004

    209. Chevalier RL, Forbes MS: Generation andevolution of atubular glomeruli in the pro-gression of renal disorders. J Am SocNephrol 19: 197206, 2008

    210. Nadasdy T, Laszik Z, Blick KE, Johnson DL,Silva FG: Tubular atrophy in the end-stagekidney: A lectin and immunohistochemicalstudy. Hum Pathol 25: 2228, 1994

    211. Docherty NG, OSullivan OE, Healy DA,Fitzpatrick JM, Watson RW: Evidence thatinhibition of tubular cell apoptosis protectsagainst renal damage and development offibrosis following ureteric obstruction.Am J Physiol Renal Physiol 290: F4F13,2006

    212. Attanasio M, Uhlenhaut NH, Sousa VH,OToole JF, Otto E, Anlag K, Klugmann C,Treier AC, Helou J, Sayer JA, Seelow D,Nurnberg G, Becker C, Chudley AE, Nurn-berg P, Hildebrandt F, Treier M: Loss ofGLIS2 causes nephronophthisis in humansand mice by increased apoptosis and fibro-sis. Nat Genet 39: 10181024, 2007

    213. Neilson EG: Plasticity, nuclear diapause,and a requiem for the terminal differentia-tion of epithelia. J Am Soc Nephrol 18:19951998, 2007

    214. Chase LG, Ulloa-Montoya F, Kidder BL,Verfaillie CM: Islet-derived fibroblast-likecells are not derived via epithelial-mesen-chymal transition from Pdx-1 or insulin-pos-itive cells. Diabetes 56: 37, 2007

    215. Taura K, Miura K, Iwaisako K, Osterreicher

    BRIEF REVIEWwww.jasn.org

    J Am Soc Nephrol 21: 18191834, 2010 Mechanisms of Tubulointerstitial Fibrosis 1831

  • CH, Kodama Y, Penz-Osterreicher M, Bren-ner DA: Hepatocytes do not undergo epi-thelial-mesenchymal transition in liver fi-brosis in mice. Hepatology 51: 10271036,2010

    216. Flier SN, Tanjore H, Kokkotou EG, Sugi-moto H, Zeisberg M, Kalluri R: Identifica-tion of epithelial to mesenchymal transitionas a novel source of fibroblasts in intestinalfibrosis. J Biol Chem 285: 2020220212,2010

    217. Zeisberg M, Yang C, Martino M, DuncanMB, Rieder F, Tanjore H, Kalluri R: Fibro-blasts derive from hepatocytes in liver fi-brosis via epithelial to mesenchymal transi-tion. J Biol Chem 282: 2333723347, 2007

    218. Tanjore H, Xu XC, Polosukhin VV, DegryseAL, Li B, Han W, Sherrill TP, Plieth D, Neil-son EG, Blackwell TS, Lawson WE: Contri-bution of epithelial-derived fibroblasts tobleomycin-induced lung fibrosis. Am J Re-spir Crit Care Med 180: 657665, 2009

    219. Kim KK, Kugler MC, Wolters PJ, RobillardL, Galvez MG, Brumwell AN, Sheppard D,Chapman HA: Alveolar epithelial cell mes-enchymal transition develops in vivo duringpulmonary fibrosis and is regulated by theextracellular matrix. Proc Natl Acad SciU S A 103: 1318013185, 2006

    220. Kim KK, Wei Y, Szekeres C, Kugler MC,Wolters PJ, Hill ML, Frank JA, BrumwellAN, Wheeler SE, Kreidberg JA, ChapmanHA: Epithelial cell alpha3beta1 integrinlinks beta-catenin and Smad signaling topromote myofibroblast formation and pul-monary fibrosis. J Clin Invest 119: 213224,2009

    221. Zeisberg EM, Potenta S, Xie L, Zeisberg M,Kalluri R: Discovery of endothelial to mes-enchymal transition as a source for carci-noma-associated fibroblasts. Cancer Res67: 1012310128, 2007

    222. Kokudo T, Suzuki Y, Yoshimatsu Y,Yamazaki T, Watabe T, Miyazono K: Snail isrequired for TGFbeta-induced endothelial-mesenchymal transition of embryonic stemcell-derived endothelial cells. J Cell Sci121: 331733124, 2008

    223. Vongwiwatana A, Tasanarong A, RaynerDC, Melk A, Halloran PF: Epithelial to mes-enchymal transition during late deteriora-tion of human kidney transplants: The roleof tubular cells in fibrogenesis. Am J Trans-plant 5: 13671374, 2005

    224. Robertson H, Ali S, McDonnell BJ, BurtAD, Kirby JA: Chronic renal allograft dys-function: The role of T cell-mediated tu-bular epithelial to mesenchymal cell tran-sition. J Am Soc Nephrol 15: 390397,2004

    225. Hertig A, Anglicheau D, Verine J, Pallet N,Touzot M, Ancel PY, Mesnard L, Brousse N,Baugey E, Glotz D, Legendre C, RondeauE, Xu-Dubois YC: Early epithelial pheno-typic changes predict graft fibrosis. J AmSoc Nephrol 19: 15841591, 2008

    226. Vitalone MJ, OConnell PJ, Jimenez-VeraE, Yuksel A, Wavamunno M, Fung CL,Chapman JR, Nankivell BJ: Epithelial-to-mesenchymal transition in early transplanttubulointerstitial damage. J Am Soc Neph-rol 19: 15711583, 2008

    227. Hildebrandt F, Attanasio M, Otto E:Nephronophthisis: Disease mechanisms ofa ciliopathy. J Am Soc Nephrol 20: 2335,2009

    228. Sun S, Ning X, Zhang Y, Lu Y, Nie Y, Han S,Liu L, Du R, Xia L, He L, Fan D: Hypoxia-inducible factor-1alpha induces Twistexpression in tubular epithelial cells sub-jected to hypoxia, leading to epithelial-to-mesenchymal transition. Kidney Int 75:12781287, 2009

    229. Zeng R, Yao Y, Han M, Zhao X, Liu XC, WeiJ, Luo Y, Zhang J, Zhou J, Wang S, Ma D,Xu G: Biliverdin reductase mediates hypox-ia-induced EMT via PI3-kinase and Akt.J Am Soc Nephrol 19: 380387, 2008

    230. Gunaratnam L, Bonventre JV: HIF in kidneydisease and development. J Am Soc Neph-rol 20: 18771887, 2009

    231. Okada H, Danoff TM, Kalluri R, Neilson EG:Early role of Fsp1 in epithelial-mesenchy-mal transformation. Am J Physiol 273:F563F574, 1997

    232. Docherty NG, OSullivan OE, Healy DA,Murphy M, ONeill AJ, Fitzpatrick JM,Watson RW: TGF-beta1-induced EMT canoccur independently of its proapoptotic ef-fects and is aided by EGF receptor activa-tion. Am J Physiol Renal Physiol 290:F1202F1212, 2006

    233. Ardura JA, Rayego-Mateos S, Ramila D,Ruiz-Ortega M, Esbrit P: Parathyroid hor-mone-related protein promotes epithelial-mesenchymal transition. J Am Soc Nephrol21: 237248, 2010

    234. Oldfield MD, Bach LA, Forbes JM, Nikolic-