view - nephrology dialysis transplantation

17
Editorial Review Treatment targets in renal fibrosis Peter Boor 1,2 , Katarı´na S ˇ ebekova´ 2 , Tammo Ostendorf 1 and Ju¨rgen Floege 1 1 Division of Nephrology, RWTH University of Aachen, Aachen, Germany and 2 Department of Clinical and Experimental Pharmacotherapy, Slovak Medical University, Bratislava, Slovakia Keywords: animal models; kidney scarring; progressive renal disease; treatment options Introduction Renal fibrosis is the principal process underlying the progression of chronic kidney disease (CKD) to end- stage renal disease (ESRD). It is a relatively uniform response involving glomerulosclerosis, tubulointersti- tial fibrosis and changes in renal vasculature (loss of glomerular and peritubular capillaries) (Figure 1). Of these, tubulointerstitial fibrosis has evolved as the most consistent predictor of an irreversible loss of renal function and progression to ESRD [1]. Mechanisms contributing to tubulointerstitial injury and tubular atrophy include glomerular proteinuria, chronic hypoxia, misdirected glomerular ultrafiltra- tion, tubular protein leakage and direct toxic insults of e.g. drugs (reviewed in detail elsewhere [1–6]) . Direct or indirect tubulointerstitial injury via oxidative stress and various effector molecules trigger cellular responses like (i) tubular epithelial cell (TEC) apopto- sis, (ii) activation of fibroblasts and their phenotypic switch to myofibroblasts, (iii) influx and/or prolifera- tion of lymphocytes/macrophages, fibrocytes (the circulating fibroblast precursors), fibroblasts as well as (iv) epithelial-to-mesenchymal transition (EMT) of TECs. Renal fibrosis provides an excellent treatment target, since a large variety of pathophysiologically distinct diseases converge finally into this single process. However, we still do not have effective therapies, nor does such a therapy exist in most other types of organ fibrosis. Why? As part of the vital repair process, the regulation and redundancy in this system must be highly effective. The consequence is an amazingly complicated process that involves many cell types and mediators [1,2,4,5,7]. Not unexpectedly, mono- therapeutic approaches, or even a combination of therapies, fail to completely stop the progression of renal fibrosis [8–10]. In addition, not all combination therapies can be additive [11]. When identifying new targets or validating potential therapeutic options, we are confronted with several problems: Rodent models often do not fully mimic the clinical situation. Apart from the obvious species differ- ences, it is often difficult to distinguish whether the tested approach truly affected the phase of inter- stitial fibrosis or whether it ameliorated the under- lying primary renal injury to an extent that halted the progression. This problem is particularly rele- vant to the present review. Few approaches have been validated in multiple models. In the experimental situation, treatment is rarely started at a time point of established fibrosis (Table 1). Different parameters and techniques are used for the evaluation of fibrosis. Often histological changes are evaluated by semi-quantitative scoring rather than objective quantitative parameters. Standardized consensus approaches are urgently required. Most studies compare intervention to no interven- tion rather than comparing different approaches amongst each other or in combination with one another. In the following, we will first discuss antifibrotic approaches, that are clinically available or close to being so, and then discuss potential new targets in fibrosis therapy (Table 2). Renin–angiotensin–aldosterone (RAAS) and kallikrein–kinin system Angiotensin-converting enzyme inhibitors (ACEI) and angiotensin II receptor type 1 blockers (ARB) are undisputedly the first line drugs in combating renal fibrosis. These drugs, however, are not able to halt the progression completely and in some conditions, like aristocholic acid-induced renal fibrosis in rats, they are Correspondence and offprint requests to: Peter Boor, MD, Division of Nephrology, RWTH University of Aachen, Pauwelsstr. 30, 52074 Aachen, Germany. Email: [email protected] Nephrol Dial Transplant (2007) 22: 3391–3407 doi:10.1093/ndt/gfm393 Advance Access publication 21 September 2007 ß The Author [2007]. Published by Oxford University Press on behalf of ERA-EDTA. All rights reserved. For Permissions, please email: [email protected] Downloaded from https://academic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Upload: others

Post on 12-Feb-2022

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: View - Nephrology Dialysis Transplantation

Editorial Review

Treatment targets in renal fibrosis

Peter Boor1,2, Katarına Sebekova2, Tammo Ostendorf1 and Jurgen Floege1

1Division of Nephrology, RWTH University of Aachen, Aachen, Germany and 2Department of Clinical and ExperimentalPharmacotherapy, Slovak Medical University, Bratislava, Slovakia

Keywords: animal models; kidney scarring;progressive renal disease; treatment options

Introduction

Renal fibrosis is the principal process underlying theprogression of chronic kidney disease (CKD) to end-stage renal disease (ESRD). It is a relatively uniformresponse involving glomerulosclerosis, tubulointersti-tial fibrosis and changes in renal vasculature (loss ofglomerular and peritubular capillaries) (Figure 1). Ofthese, tubulointerstitial fibrosis has evolved as the mostconsistent predictor of an irreversible loss of renalfunction and progression to ESRD [1].

Mechanisms contributing to tubulointerstitial injuryand tubular atrophy include glomerular proteinuria,chronic hypoxia, misdirected glomerular ultrafiltra-tion, tubular protein leakage and direct toxic insults ofe.g. drugs (reviewed in detail elsewhere [1–6]) . Director indirect tubulointerstitial injury via oxidative stressand various effector molecules trigger cellularresponses like (i) tubular epithelial cell (TEC) apopto-sis, (ii) activation of fibroblasts and their phenotypicswitch to myofibroblasts, (iii) influx and/or prolifera-tion of lymphocytes/macrophages, fibrocytes (thecirculating fibroblast precursors), fibroblasts as wellas (iv) epithelial-to-mesenchymal transition (EMT)of TECs.

Renal fibrosis provides an excellent treatment target,since a large variety of pathophysiologically distinctdiseases converge finally into this single process.However, we still do not have effective therapies, nordoes such a therapy exist in most other types of organfibrosis. Why? As part of the vital repair process, theregulation and redundancy in this system must behighly effective. The consequence is an amazinglycomplicated process that involves many cell typesand mediators [1,2,4,5,7]. Not unexpectedly, mono-therapeutic approaches, or even a combination of

therapies, fail to completely stop the progression ofrenal fibrosis [8–10]. In addition, not all combinationtherapies can be additive [11].

When identifying new targets or validating potentialtherapeutic options, we are confronted with severalproblems:

� Rodent models often do not fully mimic the clinicalsituation. Apart from the obvious species differ-ences, it is often difficult to distinguish whether thetested approach truly affected the phase of inter-stitial fibrosis or whether it ameliorated the under-lying primary renal injury to an extent that haltedthe progression. This problem is particularly rele-vant to the present review.

� Few approaches have been validated in multiplemodels.

� In the experimental situation, treatment is rarelystarted at a time point of established fibrosis(Table 1).

� Different parameters and techniques are used forthe evaluation of fibrosis. Often histologicalchanges are evaluated by semi-quantitative scoringrather than objective quantitative parameters.Standardized consensus approaches are urgentlyrequired.

� Most studies compare intervention to no interven-tion rather than comparing different approachesamongst each other or in combination with oneanother.

In the following, we will first discuss antifibroticapproaches, that are clinically available or close tobeing so, and then discuss potential new targets infibrosis therapy (Table 2).

Renin–angiotensin–aldosterone (RAAS) andkallikrein–kinin system

Angiotensin-converting enzyme inhibitors (ACEI) andangiotensin II receptor type 1 blockers (ARB) areundisputedly the first line drugs in combating renalfibrosis. These drugs, however, are not able to halt theprogression completely and in some conditions, likearistocholic acid-induced renal fibrosis in rats, they are

Correspondence and offprint requests to: Peter Boor, MD, Divisionof Nephrology, RWTH University of Aachen, Pauwelsstr. 30,52074 Aachen, Germany. Email: [email protected]

Nephrol Dial Transplant (2007) 22: 3391–3407

doi:10.1093/ndt/gfm393

Advance Access publication 21 September 2007

� The Author [2007]. Published by Oxford University Press on behalf of ERA-EDTA. All rights reserved.For Permissions, please email: [email protected]

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 2: View - Nephrology Dialysis Transplantation

not effective at all [12]. Combination therapy of ACEIand ARB [13] or high-dosage of ACEI [14] showpotential in experimental studies to arrest or evenregress renal fibrosis, at least in the early stages.

Kallikrein cleaves the precursor kininogen to theactive vasodilator kinin peptide, bradykinin, which

via its B2 receptors mediates protective effects inhypertension, renal injury and fibrosis [15,16]. Genedelivery of kallikrein ameliorated renal scarring with-out affecting the blood pressure [17], and even reversedestablished kidney fibrosis [18]. ACE is the sameenzyme as kininase II, which degrades active kinins.Thus, ACE inhibition leads to kinin accumulation,which may contribute to the beneficial effects of ACEI.

Aldosterone inhibition, e.g. with mineralocorticoidreceptor blockers (spironolacton, eplerenone), alsoshows beneficial effects in experimental as well as inclinical studies (reviewed in [19,20]).

Renin inhibitors (aliskiren, enalkiren, zalkiren) arepromising drugs for combating renal fibrosis as shownin severely hypertensive transgenic rats (dTGR)harbouring the human renin and angiotensinogengene [21]. Clinical effects on renal fibrosis remain tobe determined. The recently discovered receptor forrenin and prorenin [(P)RR – (pro)renin receptor] waslinked to glomerular fibrosis [22]. (P)RR blockersmight be even more potent than renin inhibitors, sincerenin inhibitors do not block renin or prorenin bindingto and activation of (P)RR [22].

Vasopeptidase inhibitors (e.g. AVE7688, omapatri-lat) block both ACE and neutral endopeptidase, whichresults in a more pronounced blood pressure decrease,as compared with ACEI. AVE7688 potently retardedrenal fibrosis in an animal model of Alport syndrome[23]. However, the higher incidence of angioedemawith omapatrilat as compared with ACEI has limitedclinical studies thus far.

Table 1. Examples of experimental studies that showed effective antifibrotic treatment aproaches in established secondary renal fibrosis

Experimental model Treatment Treatment duration(days after diseaseinduction)

Reducedtubulo-interstitialfibrosis (at day)

Effect onbloodpressure

Ref.

5/6 nephrectomy Enalapril via drinking water 56–84 84 Yes [14]5/6 nephrectomy MMF by daily gavage or/and losartan

via drinking water30–60 and 30–120 60, 120 Yes [47]

5/6 nephrectomy VEGF121 injections twice daily 28–56 56 No [190]5/6 nephrectomy Adenoviral delivery of DDAH

(ADMA degrading enzyme) comparedwith hydralazine

28–42 42 No [283]

Salt induced nephropathy inDahl salt-sensitive rats

Adenoviral delivery of human tissuekallikrein gene

56–70 70 Yes [18]

Spontaneously hypertensive rats Human recombinant relaxin infusion 280–294 294 Yes [82]Accelerated anti-GBM nephritis IL-1 receptor antagonist infusion 7–21 21 ND [110]Accelerated anti-GBM nephritis BMP-7 injection every other day 7–42, 14–42, 21–42

and 28–4242 ND [166]

Anti-GBM nephritis Anti-TNF-a Ab injection every other day 14–28 28 ND [123]Adriamycin nephropathy CCR1 antagonist injection every 8 h 14–42 42 ND [137]Progressive anti-Thy1.1 GN Anti-PDGF-D Ab single injections 17, 28, 35 100 No [204]Cyclosporin A nephropathy VEGF121 injections twice daily 45–59 66 Yes [191]

Only original studies cited in this review were included. Some models were not included in the table, namely those considered models ofprimary tubulointerstitial damage, i.e. UUO and Alport-mice. All studies using genetically modified animals were also not included, since thefactor is always up-regulated/absent prior to disease induction. We also excluded models of diabetes (e.g. streptozotocin-induced diabetes ordb/db mice), lupus (MRL/lpr mice or pristane-induced lupus) or of chronic allograft nephropathy, were it is not possible to discriminate,whether reduction of established fibrosis was due to amelioration of the underlying disease or the fibrosis itself.Ab, antibody; ADMA, asymmetric dimethylarginine; BMP-7, bone morphogenic protein-7; CCR, receptor for CC chemokines; DDAH,dimethylarginine dimethylaminohydrolase; IL-1, interleukin 1, MMF, mycophenolate mofetil; ND, not determined; PDGF, platelet-derivedgrowth factor; VEGF, vascular endothelial growth factor.

Fig. 1. Severe tubulointerstitial fibrosis in an experimental rat modelof progressive mesangioproliferative glomerulonephritis on day 100after disease induction. The renal cortical section stained withperiodic acid-Schiff (PAS) shows typical tubulointerstitial damage:accumulation of extracellular matrix (�), tubular atrophy (��) andinflammatory infiltrates (���). Glomerular damage is apparent asglomerulosclerosis (arrow) and proteinuria (double arrow).Magnification: 100�.

3392 P. Boor et al.

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 3: View - Nephrology Dialysis Transplantation

Endothelin, sympathic nerve system

Endothelin, acting via its receptors ETA and ETB, is apotent vasoconstrictor and mediator of fibroticresponse [2,24–26]. Although the dual inhibitor ofETA and ETB bosentan is in clinical use, its benefitsvs side effects (mainly hepatotoxicity and fluidretention) in CKD patients remain largely to beevaluated.

Blockade of sympathic nerve activity withmoxonidine reduced the progression of fibrosis in

5/6-nephrectomized rats without affecting blood pres-sure [27,28]. Selective a and b blockers in subantihy-pertensive dosages also ameliorated the developmentof renal fibrosis [29,30]. In cyclosporine-induced renaldamage, however, kidney denervation had no effect onthe development of fibrosis [31].

Environmental factors and metabolic syndrome

Smoking accelerates progression of kidney diseases,in part through blood pressure-mediated effects [32],

Table 2. Summary of reviewed targets and/or compounds for treatment of renal fibrotic disease

Target group Therapeutic target and/or approach Ref.

Renin–angiotensin–aldosterone system ACE (ACEI), Ang II R 1 (ARB), aldosterone (aldosteroneantagonists), ACE þ neutral endopeptidase(vasopeptidase inhibitors), renin (renin inhibitors,(P)RR antagonist)

[12–14,19–23]

Kallikrein–kinin system kallikrein–kinin–B2 receptors; kininase II [15–18]Endothelin ETA and ETB (dual inhibitor bosentan) [24–26]Sympathic nerve system � and � blockers, moxonidine [27–30]Environmental factors cessation of smoking, stress, caffeine intake, infections, heavy metals [32–36]Obesity, hypercholesterolaemia diet, statins [37–46]Immunosupressants MMF, rapamycin, FTY720, dexamethasone [47–56]ECM turnover

and compositionMMP-1, MMP-2, TIMPs-1, ADAM-19, ADAM-17, ADAMTS-1,

tissue transglutaminase, integrins (a1b1, aVb6), ILK, relaxin(relaxin), trypsinþbromelainþrutosid, pirfenidone

[58–65,76–83,85–92,94,95]

Fibrinolytic system PAI-1, tPA, plasminogen, uPAR [66–75]Complement system C5 (anti-C5 Ab), C5b-9 (CD59), C5a (C5aR antagonist), Crry [96–108]Cytokines IL-1 (IL-1 receptor antagonist), IL-4, IL-8, IL-10 (anti-IL-10 Ab),

IFN-g (IFN-�), IFN-a (IFN-�), TNF-a (anti-TNF-�-Ab)[109–115,

117–123,280]Chemokines MCP-1/CCR2, RANTES/CCR1, M-CSF, osteopontin, CX3CR1,

SLC/CCR7 (chemokine receptor antagonists)[127–143]

TGF-b signalling TGF-b, Smad-7, Smad-3 (halofuginone), Snail, Ski, SnoN, ALK5,decorin

[149–161,164,165]

TGF-b antagonists BMP-7, USAG-1, KCP [166–172,174,175]Other growth factors PDGF (see Table 3), HGF, CTGF (Anti-CTGF Ab), FGF-1, FGF-2,

VEGF (anti-VEGF-Ab), EGFR (anti-EGFR-Ab)[176–178,180,183–189,

192–197,212–218]Nitric oxide L-arginine, iNOS, eNOS, sGC, PDE (PDE antagonist pentoxifiline),

PDE-4, PDE-5 (PDE-5 antagonist sildenafil), NO-donors[219–232]

Intracellular transduction cascades NF-kB (curcumin), Rho/ROCK (Rho inhibitors), p38 MAPK(p38 MAPK inhibitors), JNK, PKC-b, PI3Kg (PI3K� inhibitors),transcription factor Sp1, various tyrosin kinase inhibitors

[146,161–163,233–240,273–277,281]

Various stem cells, mast cells, B-cells, selectins, AGEs, AOPPs, PPARg(glitazones), ADMA (DDAH), tranilast, 1,25-dihydroxyvitamin D,paracalcitol, retinoid receptor agonist isotretinoin, erythropoietin,polyunsaturated fatty acids, thromboxane receptor antagonistterutroban, N-acetyl-cysteine

[241,243–272,278,279,282,283]

Treatment options already available or currently being tested in humans are in italics. Intracellular transduction cascades are discussedin various parts of the text.Ab, antibody; ACE, angiotensine-converting enzyme; ACEI, ACE inhibitors; ADAM, a disintegrin and metalloprotease domain; ADAMTS,ADAM with thrombospondin motifs; ADMA, asymmetric dimethylarginine; AGE, advanced glycation end-product; ALK5, TGF-b1receptor kinase; Ang II, angiotensin II; Ang II R 1, Ang II receptor 1, AOPP, advanced oxidation protein product; ARB, angiotensin-receptor blocker; B2 receptors, bradykinine B2 receptors; BMP-7, bone morphogenic protein-7; C5a, complement component 5a; C5aR,receptor for C5a (CD88); C5b-9, membrane attack complex; CCL, CC chemokine; CCR, receptor for CC chemokines; CD59, membrane-bound complement regulatory protein; Crry, complement receptor-related protein; CTGF, connective tissue growth factor (CCN2);CX3CR1, fractalkine receptor 1; DDAH - dimethylarginine dimethylaminohydrolase; ECM, extracellular matrix; EGFR, epidermal growthfactor receptor; ETA, B, endothelin receptors A and B; FGF, fibroblast growth factor; HGF, hepatocyte growth factor; IFN, interferon; IL,interleukin; ILK, integrin linked kinase; JNK, c-Jun amino-terminal kinase; KCP, kielin/chordin-like protein; MCP-1, monocytechemoattractant protein (CCL2); M-CSF, macrophage-colony stimulating factor; MMF, mycophenolate mofetil; MMP, matrixmetalloproteinase; NF-kB, nuclear factor kB; NOS, nitric oxide synthase; p38 MAPK, p38 mitogen-activated protein kinase; PAI-1,plasminogen-activator inhibitor-1; PDE, phosphodiesterase; PDGF, platelet-derived growth factor; PDGFR, PDGF receptor; PI3Kg,phosphatidyl-inositol-3-kinase-g; PKC-b, protein kinase C-b; PPARg, peroxisome proliferator-activated receptor-g;(P)RR, (pro)reninreceptor; RANTES, regulated on activation, normal T-cell expressed and secreted (CCL5); ROCK, Rho-kinase; sGC, soluble guanylatecyclase; Ski, Sloan-Kettering Institute proto-oncogene; SLC, secondary lymphoid tissue chemokine (CCL21); SnoN, Ski-related novel gene;TGF-b, transforming growth factor-b; TIMP, tissue inhibitor of MMPs; TNF, tumor necrosis factor; tPA, tissue-type plasminogen activator;uPA, urokinase plasminogen activator; uPAR, uPA receptor; USAG-1, uterine sensitization-associated gene 1; VEGF, vascular endothelialgrowth factor.

Treatment targets in renal fibrosis 3393

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 4: View - Nephrology Dialysis Transplantation

but at least in experimental situations, also via apossible profibrotic effect [33,34]. Another indepen-dent factor that leads to progression of kidney diseaseis obesity [36]. Various pathways, including increasedmetabolic demands on the kidney, hyperinsulinaemiaor leptin overproduction, were shown to induce orpromote kidney scarring in obesity [37–40]. Diet-induced hypercholesterolaemia in rodents or pigsresulted in renal fibrosis, which was reversible bylipid-lowering dietary interventions [41,42]. Severalexperimental studies demonstrated the protectiveeffect of statins on kidney scarring, which may relateto their lipid-lowering but also to their anti-inflamma-tory actions [43–45]. First clinical data confirmed therenoprotective effects of statins [46] and large trials inCKD patients are underway.

Immunosuppressive agents

Mycophenolate mofetil (MMF) ameliorated fibrosis invarious experimental models [47–50]. These effectswere comparable, but not additive, with those ofenalapril or lisinopril [48,49]. Confirmatory data intransplanted patients has recently emerged [51].Rapamycin attenuated renal fibrosis in patients withkidney transplantation [52] and in unilateral ureteralobstruction (UUO) in rats [53]. Dexamethasonereduced fibrosis in renin transgenic rats similiarly toMMF [55], but failed to reduce fibrosis in mercuricchloride-treated rats, whereas tacrolimus was effectivein the latter [56].

Turnover and composition of the extracellular matrix

Matrix metalloproteinases (MMPs) were initiallythought to be beneficial in renal fibrosis, given thatthey degrade extracellular matrix (ECM) proteins.Recent data suggest that their role in renal scarringmay be much more complex [57] (Figure 2). In renalfibrosis, local delivery of MMP-1 reduced collagencontent in streptozotocin-induced diabetic nephropa-thy in rats [58]. On the other hand, tubular MMP-2overexpression in mice induced renal fibrosis [59] andselective pharmacological MMP-2 inhibition increasedfibrosis in UUO [60].

Tissue inhibitors of matrix metalloproteinases(TIMP-1 to -4) are natural MMP inhibitors. GeneticTIMP-1 deletion in mice had no influence on experi-mental kidney fibrosis [61,62], whereas overexpressionof TIMP-1 increased age-related fibrosis [63].

Membrane-bound ‘a disintegrin and metallopro-tease domain’ proteases (ADAMS) and secreted‘ADAM with thrombospondin motifs’ proteases(ADAMTS) are two other important families ofproteases (containing more than 50 members).ADAM-19 is abundant in fibrotic renal lesions inhuman biopsies [64]. ADAMTS-1-deficient miceexhibited kidney fibrosis in addition to renalmalformations [65].

Some proteases of the fibrinolytic pathway, i.e.plasminogen activator inhibitor-1 (PAI-1) [66–69],tissue-type plasminogen activator (tPA) [70,71] andplasminogen [72,73] are important profibrotic factors.However, in anti-glomerular basement membrane(anti-GBM) nephritis, all three factors proved to beantifibrotic, which may relate to their involvement inthe glomerular coagulation cascade, i.e. effects on theprimary renal disease as opposed to direct effects onrenal fibrosis [68,74,75]. Genetic urokinase-type plas-minogen activator (uPA) deficiency had no effect onfibrosis in UUO or anti-GBM nephritis; deficiency ofits receptor (uPAR) was antifibrotic in UUO but not inanti-GBM nephritis [66].

Tissue transglutaminase (Tg) stabilizes ECM byprotein cross-linking. This leads, for example, toresistance of such modified collagens to proteolyticdegradation by MMPs. Tissue Tg is upregulated andcorrelates closely with the severity of renal fibrosis inexperimental models and in humans [76,77]. At leastin vitro, inhibition of tissue TG reduced glucose-induced deposition of ECM proteins in renal proximaltubular epithelial cells [77].

Oral administration of a protease mixture (trypsinand bromelain with rutosid added as an antioxidant)reduced renal fibrosis in rats with 5/6-nephrectomy orGoldblatt hypertension independently of blood pres-sure [78,79]. Single proteases or their mixtures are usedin human medicine.

Relaxin is a small peptide hormone with potentantifibrotic activity in the kidney [80–82]. Its benefitmay derive in part from interference with transforminggrowth factor-b (TGF-b), a potent profibrotic cytokine[83]. Continuous subcutaneous infusion of relaxinshowed promising results in a phase II clinical trial inpatients with systemic sclerosis, but this effect was notconfirmed in a phase III trial [84].

Pirfenidone is a potent inhibitor of ECM accumula-tion as shown in several experimental models ofrenal damage, e.g. mesangioproliferative GN,5/6-nephrectomy or UUO [85,86], and in patientswith idiopathic pulmonary fibrosis and advanced liverfibrosis. We lack clinical data in renal patients.

Integrins are heterodimeric cell receptors for theECM. Integrin a1 chain-deficient mice developed moresevere glomerulosclerosis in adriamycin-induced renalinjury [87]. In contrast, genetic a1 chain-deficiency orantagonism of a1 chain reduced fibrosis in a mousemodel of Alport syndrome [88] and in rats withcrescentic GN [89] or mesangioproliferative GN [90].Integrin aVb6 binds to and activates latent TGF-b.Mice deficient of the b6 integrin chain exhibitedreduced fibrosis following UUO [91]. Deficiency ofb6 or aVb6-blocking antibodies also reduced fibrosisin Alport mice [92]. In contrast, genetic deficiency ofthe a8-chain did not contribute to glomerulosclerosisor fibrosis [93]. Outside-in signalling of integrinsinvolves, amongst other functions, activation of theintegrin-linked kinase (ILK) pathway. ILK wassuggested to participate in the development of renalfibrosis [94,95].

3394 P. Boor et al.

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 5: View - Nephrology Dialysis Transplantation

Collectively these data suggest that some systems,such as MMPs, the fibrinolytic pathway and integrins,may be difficult to interfere with clinically, given theircomplex nature, their redundancy and their sometimesopposing biological effects. However, some attractivetherapeutic options such as Tg- antagonists are startingto emerge.

Complement system

The terminal complex of complement, C5b-9 (ormembrane attack complex—MAC) is formed at sitesof tubulointerstitial injury [1], its depletion in experi-mental nephropathy reduces proteinuria [96] andinhibition of MAC formation in proteinuric animal

models ameliorated tubulointerstitial injury [1,97–99].However, genetic C6 deficiency, which precludesMAC formation, did not affect the severity of non-proteinuric models of tubulointerstitial injury [100],suggesting that MAC mediates tubulointerstitialdamage in proteinuric renal diseases only [100].

Besides C5b-9, complement C5 and in particularC5a may also become a target in renal fibrosis: inexperimental murine immune-complex GN, geneticdeficiency of the C5a receptor (C5aR) led to reducedinterstitial cell infiltration and tubulointerstitialdamage without affecting the glomerular injury. Thispointed to a role of the anaphylatoxin C5a, a smallpeptide released from C5, in tubulointerstitial injury[105]. In the mouse UUO model, genetic C5 deficiencyor pharmacological inhibition of C5aR potently

Fig. 2. Complexity of MMP driven proteolysis. Each of the 23 human and 24 murine MMPs can be regulated at different levels, ranging fromRNA processing to activation of inactive MMPs (zymogens) and/or degradation of active MMPs by other proteases (e.g. other MMPs). Thebroad range of the MMPs targets resulting in a diversity of biological functions adds another level of complexity to this protease system. Withrespect to fibrosis, it seems obvious, that MMPs should not be viewed as merely ECM degrading ‘anti-fibrotic’ molecules. Adapted andmodified from [285]. ECM, extracellular matrix; EGFR, epidermal growth factor receptor; EMT, epithelial-to-mesenchymal transition; FGF,fibroblast growth factor; IL-8, interleukin-8; MCP-1, monocyte chemoattractant protein 1; MMP, matrix metalloproteinase; PDGF, platelet-derived growth factor; PDGFR, PDGF receptor; TGF-b, transforming growth factor-b; TIMP, tissue inhibitor of MMPs; TNF-a, tumournecrosis factor-a; VEGF, vascular endothelial growth factor.

Treatment targets in renal fibrosis 3395

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 6: View - Nephrology Dialysis Transplantation

reduced tubulointerstitial fibrosis [106]. C5aR defi-ciency or antagonism also attenuated the course oflupus nephritis in mice [107,108]. C5aR antagonistsand anti-C5 antibodies (eculizumab) were/are beingevaluated in clinical trials.

Complement regulatory proteins such as Crry, whichinhibits C3 activation, or CD59, which inhibits C5b-9formation, may also be of interest: Local deficiencyof Crry aggravated tubular damage and fibrosis inpuromycin-induced nephropathy (PAN) and in modelsof renal transplantation [101,102]. Crry and CD59were equally therapeutic in PAN [103]. Treatment withCrry also decreased renal ECM accumulation inmurine lupus nephritis [104], but here, effects on theimmune system vs direct, antifibrotic effects aredifficult to distinguish.

Cytokines

Few data are available on the role of interleukins (IL)in renal fibrosis. One exception is IL-1b, whoseprofibrotic properties in experimental renal diseasehave been established [109–113]. Although an IL-1receptor antagonist (anakinra) is already in clinical use,it has not been tested so far in patients with renalfibrosis. In mice lacking the IL-8 receptor, whichmediates transepithelial migration, trapped neutrophilslead to tissue destruction and renal fibrosis afterexperimental pyelonephritis [114]. An antifibroticaction of IL-10 was described in 5/6-nephrectomizedrats [115]. IL-10 is also anti-inflammatory and anti-atherogenic and systemic treatment was well toleratedin psoriasis patients [116]. However, systemic treat-ment with IL-10 was ineffective in patients withrheumatoid arthritis or Crohn’s disease [116], unlesslocally delivered in the gut of Crohn’s patients. Thus,local delivery in renal patients could become a limitingfactor. It also should be stressed that in systemic lupuserythematosus IL-10 seems to be a harmful factor[116]. Early IL-4 administration in rats with crescenticGN diminished tubulointerstitial fibrosis in the latercourse of the disease [117]. Although IL-5, -6 and -13are involved in the pathogenesis of some renal diseases,no data currently exist on their role in renal scarring.

Interferon-g (IFN-g) has antifibrotic propertiesin vitro [118] and administration to 5/6-nephrectomizedrats decreased kidney fibrosis [119]. However, in ratswith mesangiopoliferative GN, IFN-g treatmentdecreased mesangial cell proliferation but not glomer-ular ECM accumulation [120]. IFN-a reduced intersti-tial fibrosis in carbon tetrachloride-inducednephrotoxicity [121]. Although IFN treatment long-term clinical therapy is feasible, the case for such treat-ment in renal fibrosis patients is not particularly strong.

Tumour necrosis factor-a (TNF-a) is a well-recognized promoter of fibrosis in the kidney[122–124]. Although anti-TNF-a antibodies arehighly effective in patients with rheumatoidarthritis, side effects such as serious infections andmalignancies [125], or increased mortality in patients

with chronic heart failure [126], may limit clinical trialsin renal fibrosis.

Chemokines

Several chemokines and their receptors were shownto act in a profibrotic manner, mainly via recruitmentof inflammatory cells into the tubulointerstitium [6].These profibrotic molecules include monocyte chemoa-tractant protein-1 (MCP-1/CCL2) and its receptorCCR2 [127–133], RANTES (CCL5) and its receptorCCR1 (but not CCR5) [134–137], macrophage-colonystimulating factor (M-CSF) [138], osteopontin[139–141] and fractalkine receptor 1 (CX3CR1) [142].The secondary lymphoid tissue chemokine(SLC/CCL21) and its receptor CCR7 have beenimplicated in the regulation of fibrocyte infiltrationof the renal interstitium and promotion of fibrosis[143]. Several problems render a translation of theseresults into clinical trials difficult [6]: (i) There areconsiderable species differences in the expression ofchemokines and their receptors. (ii) Although profi-brotic in most disease models, chemokines might bealso beneficial; e.g. lack of CCR1 or CCR2 wasantifibrotic in UUO but aggravated renal injury increscentic GN [144,145]. (iii) Chemokine receptorantagonists developed for clinical use, e.g. antagonistsof CCR2 and CCR5, cross-react with other closelyrelated G-protein-coupled receptors, resulting in insuf-ficient specificity in humans. An alternative approachcould be the inhibition of chemokine signalling viaphosphatidylinositol-3-kinase-g (PI3Kg). Indeed,a PI3Kg inhibitor ameliorated the severity of lupusnephritis in mice [146].

TGF-�, TGF-� signalling molecules, p38 MAPK,bone morphogenic protein-7 (BMP-7)

TGF-b, a central mediator of fibrosis, exerts itsbiological, in particular immunological, functions viacomplex signalling pathways [147]. In view of its potentaction as an endogenous immunosuppressant, com-plete blockade of TGF-b signalling could have seriousadverse consequences. Thus, TGF-b-deficient micedie of a multifocal inflammatory syndrome [148].However, the complexity of TGF-b regulation and itsdownstream pathways provide possibilities for morespecific treatment targets (e.g. BMP-7, HGF, CTGF,Smads) (Figure 3). It should be mentioned that whilemost of the research focuses on the TGF-b1 isoform,the other isoforms, b2 and b3, also have profibroticeffects on kidney cells [149].

Overexpression of Smad7, an inhibitory factor in theTGF-b signalling pathway, reduced renal fibrosis in5/6-nephrectomy and UUO [150–152]. Alternatively,genetic deletion of the agonistic signalling molecule,Smad3, protected kidneys from interstitial fibrosis[153,154]. The plant alkaloid halofuginone is aninhibitor of collagen I synthesis, recently shown toalso inhibit Smad3 [155]. Halofuginone was

3396 P. Boor et al.

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 7: View - Nephrology Dialysis Transplantation

successfully used for topical treatment of fibrosis inpatients with scleroderma, had good tolerability afteroral administration and is currently being tested incancer patients. Snail is a transcription factor involvedin EMT and is strongly activated by TGF-b.Activation of Snail in adult transgenic mice inducedrenal fibrosis and Snail was found to be overexpressedin human fibrotic kidney [156]. Other targets in renalscarring might be the antifibrotic Smad co-repressors,Ski and SnoN [157,158]. Inhibition of the TGF-b1receptor kinase (ALK5) alone [159,160] or in combina-tion with inhibition of p38 mitogen-activated proteinkinase (MAPK) [161] ameliorated renal fibrosis.Inhibition of p38 MAPK also reduced fibrosis inUUO and unilateral kidney ischaemia–reperfusion[162,163], and p38 MAPK inhibitors are currentlytested in clinical trials in patients with inflammatory

diseases, e.g. asthma. Lack of decorin, a proteoglycaninvolved in ECM assembly and a TGF-b antagonist,elevated interstitial fibrosis in UUO [164] and deliveryof decorin by gene therapy prevented fibrosis inglomerulonephritic rats [165].

BMP-7 was shown to reduce (more potently thanenalapril) or even reverse renal interstitial fibrosis invarious experimental models [166–170]. Furthermore,treatment with BMP-7 had beneficial effects on renalosteodystrophy and reduced vascular calcification inuraemia [171,172]. However, in the protein overloadmodel in rats, treatment with BMP-7 protein showedno significant effects on renal fibrosis [173]. BMP-7therapy in patients has not yet been reportedbut the results of ongoing studies are eagerlyawaited. Kielin/chordin-like protein (KCP) and uterinesensitization-associated gene-1 (USAG-1) are an endo-genous BMP-7 agonist and antagonist, respectively.Mice lacking KCP were more susceptible to develop-ment of renal fibrosis [174], whereas mice lackingUSAG-1 were protected [175].

Hepatocyte growth factor (HGF) and connectivetissue growth factor (CTGF; CCN2)

HGF was shown to have antifibrotic properties indifferent animal models via inhibitory effects onSmad2/3 and activation of SnoN, which binds andinactivates Smad2 [176–179]. However, HGF over-expression or its glomerular ultrafiltration in diabeticnephropathy rats with proteinuria have also beenimplicated in the development of renal fibrosis[180,181]. The pro-carcinogenic HGF effects [182]raise safety concerns for clinical trials.

CTGF, a potent profibrotic molecule, is at least inpart a direct downstream mediator of TGF-b. Itdecreases vascular endothelial growth factor (VEGF)signalling, enhances signalling of several growthfactors [e.g. TGF-b itself, epidermal growth factor(EGF), platelet-derived growth factor (PDGF), basicfibroblast growth factor (bFGF)] and directly regulatescell functions (adhesion, migration, proliferation) viabinding to integrins and ECM [183,184]. CTGF isa direct mediator of profibrotic effects of AGEs,is up-regulated in humans with renal fibrosis and itsinhibition resulted in a potent reduction of fibrosisin experimental diabetic nephropathy, UUO and5/6-nephrectomy [183–188]. Anti-CTGF therapy isnow in clinical trials in diabetic nephropathy.

Vascular endothelial growth factor (VEGF)

VEGF is a potent angiogenic factor, which isalso involved in fibrosis [189]. In rats with 5/6-nephrectomy, cyclosporine nephropathy and throm-botic microangiopathy, administration of VEGF121

reduced fibrosis and renal damage [189–191].Consistent with this, VEGF antagonism acceleratedrenal damage in mice with progressive crescentic GN[192] and in rats with mesangioproliferative GN [193].

Fig. 3. TGF-b signalling and it modulation. Thesimplified scheme shows Smad signalling of TGF-b and BMP-7and mainly encompasses molecules discussed in the text. Differentmolecules modify extracellularly the activity of TGF-b or BMP-7(note: CTGF effects on BMP-4 have been shown, but its effects onBMP-7 signalling are less clear). TGF-b or BMP-7 binding to theirreceptors induces heteromeric receptor complexes with kinaseactivation (ALK5 and ALK3, respectively) that leads to recruitmentand phosphorylation of Smads (Smad2/3 and Smad1/5, respec-tively). These phospho-Smads (pSmad) form heteromers with Smad4and are transported to the nucleus where they regulate geneexpression. Antagonistic effects of BMP-7 on TGF-b signalling aredepicted in the example of collagen type I expression and onnuclear shuttling and phosphorylation of Smad3. Non-SmadTGF-b signalling, shown in the right side of the picture, can involveactivation of p38 MAPK, JNK and Rho. The modulationof TGF-b-induced gene transcription by Smad co-repressorsSki and SnoN in nucleus is also shown. Arrows indicatestimulatory effects and blunted lines inhibitory effects. ‘P’ standsfor phosphorylation.ALK3, BMPR-IA kinase; ALK5, TbRI kinase; BMP-7, bonemorphogenic protein-7; BMPR-IA, BMP receptor IA; BMP-RII,BMP receptor II; JNK, c-Jun amino-terminal kinase; KCP, kielin/chordin-like protein; p38 MAPK, p38 mitogen-activated proteinkinase; TbR, TGF-b1 receptor; TGF-b, transforming growthfactor-b; USAG-1, uterine sensitization-associated gene 1.

Treatment targets in renal fibrosis 3397

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 8: View - Nephrology Dialysis Transplantation

However, the action of VEGF may depend on thebiological context, since, in contrast to the above data,a VEGF neutralizing antibody ameliorated early renalinjury, in particular glomerular hypertrophy, in5/6-nephrectomized rats, in mice fed with a high-protein diet and in rats with diabetic nephropathy[194–197]. Various anti-VEGF aproaches are approvedfor use in patients, and renal side effects includeproteinuria and hypertension [198]. Given the abovedata, the complexity of the VEGF system (five familymembers with alternative splicing, four differentreceptors) and potential adverse pro-angiogenic effectsof VEGF-therapy, it appears uncertain whether VEGFadministration or blockade will become a usefulapproach in patients with renal fibrosis.

Platelet-derived growth factor (PDGF)

PDGF is a key mediator of fibrosis in different organs,including the kidney (Table 3 and Figure 4). ThePDGF–B and –D isoforms mediate glomerular ECMaccumulation in GN [199]. Their main receptor, i.e.PDGFR-b (Figure 4), is upregulated in the fibrotictubulointerstitium [199]. PDGF-B also exerts itsprofibrotic effects in the tubulointerstitial compart-ment [200,201]. PDGF-D expression increases inobstructive uropathy in both humans and mice [202].In rats with progressive mesangioproliferative GN,treatment with PDGF-D-neutralizing antibody ame-liorated the early glomerular damage and the sub-sequent tubulointerstitial fibrosis [203]. Furthermore,this treatment prevented progression and fibrosis, evenif initiated at the stage of established tubulointerstitialdamage [204]. In a phase I clinical trial, a PDGF-D-neutralizing antibody had promising safety/tolerabilityprofile, pharmacodynamic properties and long half-life(Hahne et al., J Am Soc Nephrol 2005; 16: AbstractSA-PO1017).

The PDGFR-a ligand PDGF-C (Figure 4), alsoseems to be an important mediator of fibrogenic

stimuli. PDGF-C is expressed de novo in the fibroticinterstitium of rat and human kidneys [205,206] and itsspecific antagonism in the mouse UUO reduced fibroticchanges (Eitner et al., submitted for publication).

Imatinib (STI-571) is widely used in cancer therapyas a multi-kinase blocker of the c-abl, c-kit andPDGFR tyrosine kinases. Imatinib ameliorated renalfibrosis in UUO [207] and retarded the developmentof experimental diabetic [208] and chronic allograftnephropathy [209]. It remains uncertain whether thisbeneficial effect of imatinib was indeed mediated viareduction of PDGF signalling or whether it related toeffects on c-abl kinase [207]. In clinical studies, sideeffects of imatinib include effects on haematopoesis,heart [210] and bone [211].

Other growth factors

The role of basic fibroblast growth factor-2 (FGF-2)in renal fibrosis is well established, at least in vitro[212–214]. FGF-1 and its receptor were also identifiedin human interstitial fibrotic lesions [215]. Specificinterventions have not been performed in either casein renal fibrosis.

Inhibition of epidermal growth factor receptor(EGFR) is protective against the development offibrosis in models of hypertensive renal damage andrenal mass reduction [216,217]. Expression of adominant negative EGFR prevented renal lesionsinduced by chronic Ang II infusion [218].

Similarly protective in the above model were thegenetical deletion of TGF-a or pharmacologicalinhibition of its activating sheddase TACE(ADAM17) [218]. Anti-EGFR treatment is approvedfor treatment of some cancers.

Nitric oxide, NF-�B and Rho/Rho kinase

Enhanced production of nitric oxide (NO) followingL-arginine administration prevented progression of

Table 3. Experimental in vivo modulation of PDGF signalling in renal fibrosis

Target Experimental model Aproach Fibrosis Ref.

PDGF-A normal rat PDGF-A administration none [201]PDGF-B normal rat PDGF-B administration " [201]

normal mice Hepatic overexpression " [284]rat progressive anti-Thy1 GN Aptamer antagonist # [200]

PDGF-C UUO Antibody # unpub.UUO Knock-out mice # unpub.

PDGF-D normal mice Hepatic overexpression " [284]rat progressive anti-Thy1.1 GN Antibody # [203]rat progressive anti-Thy1.1 GN Antibody # [204]

PDGFR UUO Imatinib # [207]tyrosine diabetic nephropathy Imatinib # [208]kinase chronic allograft nephropathy Imatinib # [209]

Imatinib is a multikinase inhibitor and its effect might also be contributed to inhibition of other kinases, e.g. c-abl kinase (discussed in text).# decreased fibrosis; " induced fibrosis.GN, glomerulonephritis; PDGF, platelet-derived growth factor; PDGFR, PDGF receptor; unpub., unpublished data; UUO, unilateralureteral obstruction.

3398 P. Boor et al.

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 9: View - Nephrology Dialysis Transplantation

renal fibrosis in several models [219–221] but acceler-ated it in mice with lupus nephritis [222] andadriamycin nephropathy [223]. In two differentmodels of non-immune progressive kidney damage,

pharmacological inhibitors of inducible NO synthase(NOS) [223] or endothelial NOS [224] reduced renalfibrosis. Studies in the UUO model using iNOS-deficient mice or using liposome-mediated iNOS genetransfer confirmed the protective role of iNOS[225,226], and genetic eNOS deficiency in mice led toprogressive focal kidney scarring [227]. A secondmessenger of NO, cGMP, is synthesized by solubleguanylate cyclase (sGC) and degraded by phospho-diesterases (PDE). Stimulation of sGC slowed fibrosisdevelopment in progressive mesangioproliferative GN[228]. The PDE-5 inhibitor, sildenafil, effectivelyprevented the developmet of fibrosis, but it wasineffective in reversing established lesions [229].Another PDE inhibitor, pentoxifiline, was antifibroticin UUO and, combined with an ACEI, after 5/6nephrectomy [230,231], but was less effective in ahead-to-head comparison with a compound stimulat-ing sGC [231]. In UUO, however, neither PDE-4inhibition nor an A2A adenosine receptor agonist wereable to reduce kidney damage [232].

Nuclear factor-kB (NF-kB) activation is a centralconvergence point of many proinflammatory pathwaysand a downstream effector of profibrotic moleculessuch as AGEs, the RAAS system or Smad7 [233].Reduced renal fibrosis was noted following inhibitionof NF-kB, e.g. with curcumin (diferuloylmethane)[233,234]. First clinical trials with curcumin in patientswith malignancies showed no dose-dependent toxicityand phase II trials are underway.

Signalling via the G protein Rho and its associatedRho-kinase (ROCK) has been linked with renalfibrosis using two Rho-kinase inhibitors, Y-27632and fasudil [235–238]. Both were already successfullyused in clinical trials in patients with cardiovasculardiseases [239]. Nevertheless, genetic deletion ofROCK1 had no effect on renal fibrosis in UUO[240]. The involvement of Rho/ROCK in human renaldisease is largely unkown.

Stem cells

Stem cells hold great promise in acute renal failure andpossibly even in chronic, fibrosing renal failure [241].Indeed, three recent studies showed a beneficial effectof bone marrow-derived cells in a mice model of Alportsyndrome [242–244]. However, very few studies haveso far addressed the latter situation, and the challengeis to prevent stem cells receiving profibrotic signalsfrom their environment and/or to prevent theirmaldifferentiation. Thus, it has recently been shownthat bone marrow-derived myofibroblasts will con-tribute to renal fibrosis following ischaemia reperfu-sion injury [245]; and we observed thatadministration of mesenchymal stem cells to ratswith progressive mesangioproliferative GN initiallyimproved acute renal failure, but in the long-term,these cells adopted an adipocyte-like phenotypein glomeruli and induced an intense fibroticresponse [246].

Fig. 4. Simplified scheme of PDGF signalling and its regulation.Only the main molecules relevant for this review are listed.PDGF -AA, -AB and -BB are secreted in an active form whereasPDGF -DD and -CC are activated extracellularly via proteolyticcleavage of CUB-domain by urokinase plasminogen activator (tPA)or tissue plasminogen activator (uPA), respectively. Binding ofPDGF isoforms results in autophosphorylation of the appropriatePDGF receptor. This subsequently leads to recruitment andactivation of downstream signalling pathways such as Jak/STAT,PI3K, PLC-g or MAPK, which via regulation of gene expressionmediate the biological functions of PDGF isoforms, e.g. prolifera-tion, chemotaxis, migration or ECM production. The signallingpathways depicted are common to all three PDGF receptors and thesignal transduction results in overlapping yet distinct biologicaleffects depending on the receptor and cell type (e.g. via differentbinding affinities of the signalling molecules to phosphorylatedreceptors). An example of a relevant profibrotic cross-talk is thecooperation of PDGF and integrin signalling. Binding of ECMcomponents (e.g. fibronectin, laminin) to integrins activatesoutside-in signalling via the focal adhesion kinase (FAK) pathwaythat enhances PDGF induced MAPK signalling. This results inenhanced cell proliferation and migration. Integrins also cooperatewith other receptors for growth factors and directly activatesignalling pathways such as PI3K, JNK and ILK. Various otherregulatory steps might be involved in PDGF-signalling in fibrosis,e.g. regulation of PDGF and PDGFR expression, PDGF degrada-tion or binding to non-signalling molecules (e.g. SPARC or heparin-sulphate proteoglycans). ECM, extracellular matrix; FAK, focaladhesion kinase; ILK, integrin linked kinase; JAK, Janus kinases;JNK, c-Jun amino-terminal kinase; MAPK, mitogen-activatedprotein kinase; PDGF, platelet-derived growth factor; PDGFR,PDGF receptor; PI3K, phosphatidyl-inositol-3-kinase; PLC-g,phospholipase C-g; STAT, Signal transducers and activators oftranscription; tPA, tissue-type plasminogen activator; uPA, uroki-nase plasminogen activator.

Treatment targets in renal fibrosis 3399

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 10: View - Nephrology Dialysis Transplantation

Other treatment targets and approaches

There is a number of targets that might be potentiallyeffective in treatment of renal fibrosis (Table 2). Someof them were already tested in several experimentaland/or small clinical studies, e.g. tranilast, 1,25dihydroxyvitamin D, erythropoietin or glitazones,whereas others are single-study reports or descriptivestudies without functional links to renal fibrosis(Table 2).

Conclusions

In the past years, many promising targets for thetreatment of renal fibrosis have been validated invarious animal models, and even more new targetshave been identified. For several of the targetsreviewed, substances/drugs have already been devel-oped, are being tested, or are already being therapeu-tically employed in patients with non-renal indications.Renal fibrosis, in contrast, remains a largely unchartedterritory in clinical trials. The reasons for this arecertainly multifactorial and may include long studydurations if hard endpoints, i.e. loss of GFR, are to beaimed for and, in particular, the lack of non-invasivemarkers or diagnostic tools to assess kidney scarring,and thus, monitor therapy. However, the industry hasnoted the enormous potential market, given thepossibility of developing antifibrotic therapy thatmight be of benefit in many different types of organfibrosis. Furthermore, there is hope that with a largeconsortia search for biomarkers and advancing ultra-sound, or through MR-based or molecular-imagingtechniques, even monitoring of the disease process maybecome feasible in the near future.

Acknowledgements. We apologize to all authors whose importantwork we could not cite due to space limitations. This work wassupported by grants from the Deutsche Forschungsgemeinschaft(DFG) OS 196/1-1 (T.O.) and SFB 542, project C7 (T.O., J.F.).

Conflict of interest statement. None declared.

References

1. Nangaku M. Mechanisms of tubulointerstitial injury in thekidney: final common pathways to end-stage renal failure.Intern Med 2004; 43: 9–17

2. Chatziantoniou C, Dussaule JC. Insights into the mechanisms ofrenal fibrosis: is it possible to achieve regression? Am J PhysiolRenal Physiol 2005; 289: F227–F234

3. Eddy AA. Molecular basis of renal fibrosis. Pediatr Nephrol 2000;15: 290–301

4. Iwano M, Neilson EG. Mechanisms of tubulointerstitial fibrosis.Curr Opin Nephrol Hypertens 2004; 13: 279–284

5. Liu Y. Renal fibrosis: new insights into the pathogenesis andtherapeutics. Kidney Int 2006; 69: 213–217

6. Sean EK, Cockwell P. Macrophages and progressive tubulointer-stitial disease. Kidney Int 2005; 68: 437–455

7. Eitner F, Floege J. Novel insights into renal fibrosis. Curr OpinNephrol Hypertens 2003; 12: 227–232

8. Kondo S, Shimizu M, Urushihara M et al. Addition of theantioxidant probucol to angiotensin II type I receptor antagonistarrests progressive mesangioproliferative glomerulonephritis in

the rat. J Am Soc Nephrol 2006; 17: 783–7949. Yu L, Border WA, Anderson I, McCourt M, Huang Y,

Noble NA. Combining TGF-beta inhibition and angiotensin IIblockade results in enhanced antifibrotic effect. Kidney Int 2004;

66: 1774–178410. Yang J, Dai C, Liu Y. Hepatocyte growth factor gene therapy

and angiotensin II blockade synergistically attenuate renalinterstitial fibrosis in mice. J Am Soc Nephrol 2002; 13:2464–2477

11. El Chaar M, Chen J, Seshan SV et al. The effect of combination

therapy with enalapril and the TGF-{beta} antagonist 1D11 inunilateral ureteral obstruction. Am J Physiol Renal Physiol 2007;292: F1291–F1301

12. Debelle FD, Nortier JL, Husson CP et al. The renin-angiotensin

system blockade does not prevent renal interstitial fibrosisinduced by aristolochic acids. Kidney Int 2004; 66: 1815–1825

13. Wolf G, Ritz E. Combination therapy with ACE inhibitors andangiotensin II receptor blockers to halt progression of chronic

renal disease: pathophysiology and indications. Kidney Int 2005;67: 799–812

14. Adamczak M, Gross ML, Krtil J et al. Reversal of glomerulo-sclerosis after high-dose enalapril treatment in subtotally

nephrectomized rats. J Am Soc Nephrol 2003; 14: 2833–284215. Bledsoe G, Crickman S, Mao J et al. Kallikrein/kinin protects

against gentamicin-induced nephrotoxicity by inhibition ofinflammation and apoptosis. Nephrol Dial Transplant 2006; 21:

624–63316. Schanstra JP, Neau E, Drogoz P et al. In vivo bradykinin B2

receptor activation reduces renal fibrosis. J Clin Invest 2002; 110:371–379

17. Chao J, Bledsoe G, Yin H, Chao L. The tissue kallikrein-kinin

system protects against cardiovascular and renal diseases andischemic stroke independently of blood pressure reduction. BiolChem 2006; 387: 665–675

18. Bledsoe G, Shen B, Yao Y, Zhang JJ, Chao L, Chao J.Reversal of renal fibrosis, inflammation, and glomerular

hypertrophy by kallikrein gene delivery. Hum Gene Ther 2006;17: 545–555

19. Hostetter TH, Ibrahim HN. Aldosterone in chronic kidney andcardiac disease. J Am Soc Nephrol 2003; 14: 2395–2401

20. Hollenberg NK. Aldosterone in the development and progres-

sion of renal injury. Kidney Int 2004; 66: 1–921. Pilz B, Shagdarsuren E, Wellner M et al. Aliskiren, a human

renin inhibitor, ameliorates cardiac and renal damage in double-transgenic rats. Hypertension 2005; 46: 569–576

22. Nguyen G. The (pro)renin receptor: pathophysiological roles in

cardiovascular and renal pathology. Curr Opin NephrolHypertens 2007; 16: 129–133

23. Gross O, Koepke ML, Beirowski B, Schulze-Lohoff E,Segerer S, Weber M. Nephroprotection by antifibrotic and

anti-inflammatory effects of the vasopeptidase inhibitorAVE7688. Kidney Int 2005; 68: 456–463

24. Hocher B, Paul M. Transgenic animal models for the analysis ofthe renal endothelin system. Nephrol Dial Transplant 2000; 15:

935–93725. Neuhofer W, Pittrow D. Role of endothelin and endothelin

receptor antagonists in renal disease. Eur J Clin Invest 2006; 36

[Suppl 3]: 78–8826. Pfab T, Thone-Reineke C, Theilig F et al. Diabetic endothelin B

receptor-deficient rats develop severe hypertension

and progressive renal failure. J Am Soc Nephrol 2006; 17:

1082–108927. Amann K, Rump LC, Simonaviciene A et al. Effects of low dose

sympathetic inhibition on glomerulosclerosis and albuminuria

in subtotally nephrectomized rats. J Am Soc Nephrol 2000; 11:

1469–1478

3400 P. Boor et al.

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 11: View - Nephrology Dialysis Transplantation

28. Vonend O, Apel T, Amann K et al. Modulation of geneexpression by moxonidine in rats with chronic renal failure.Nephrol Dial Transplant 2004; 19: 2217–2222

29. Amann K, Koch A, Hofstetter J et al. Glomerulosclerosis andprogression: effect of subantihypertensive doses of alpha and

beta blockers. Kidney Int 2001; 60: 1309–132330. Pawluczyk IZ, Patel SR, Harris KP. The role of the alpha-1

adrenoceptor in modulating human mesangial cell matrixproduction. Nephrol Dial Transplant 2006; 21: 2417–2424

31. Elzinga LW, Rosen S, Burdmann EA, Hatton DC, Lindsley J,

Bennett WM. The role of renal sympathetic nerves in experi-mental chronic cyclosporine nephropathy. Transplantation 2000;69: 2149–2153

32. Orth SR. Smoking and the kidney. J Am Soc Nephrol 2002; 13:

1663–167233. Odoni G, Ogata H, Viedt C, Amann K, Ritz E, Orth SR.

Cigarette smoke condensate aggravates renal injury in the renalablation model. Kidney Int 2002; 61: 2090–2098

34. Elliot SJ, Karl M, Berho M et al. Smoking induces glomerulo-

sclerosis in aging estrogen-deficient mice through cross-talkbetween TGF-beta1 and IGF-I signaling pathways. J Am SocNephrol 2006; 17: 3315–3324

35. Bennett WM, Walker RG, Henry JP, Kincaid-Smith P.

Chronic interstitial nephropathy in mice induced by psy-chosocial stress: potentiation by caffeine. Nephron 1983; 34:110–113

36. Hsu CY, McCulloch CE, Iribarren C, Darbinian J, Go AS.

Body mass index and risk for end-stage renal disease. Ann InternMed 2006; 144: 21–28

37. Abrass CK. Lipid metabolism and renal disease. ContribNephrol 2006; 151: 106–121

38. Kramer H. Obesity and chronic kidney disease. Contrib Nephrol2006; 151: 1–18

39. Lavaud S, Poirier B, Mandet C et al. Inflammation is pro-

bably not a prerequisite for renal interstitial fibrosis innormoglycemic obese rats. Am J Physiol Renal Physiol 2001;280: F683–F694

40. Wolf G, Ziyadeh FN. Leptin and renal fibrosis. Contrib Nephrol

2006; 151: 175–8341. Chade AR, Mushin OP, Zhu X et al. Pathways of renal fibrosis

and modulation of matrix turnover in experimental hypercho-lesterolemia. Hypertension 2005; 46: 772–779

42. Joles JA, Kunter U, Janssen U et al. Early mechanisms of renal

injury in hypercholesterolemic or hypertriglyceridemic rats.J Am Soc Nephrol 2000; 11: 669–683

43. Li C, Sun BK, Lim SW et al. Combined effects of losartan andpravastatin on interstitial inflammation and fibrosis in chronic

cyclosporine-induced nephropathy. Transplantation 2005; 79:1522–1529

44. Li C, Yang CW, Park JH et al. Pravastatin treatment attenuatesinterstitial inflammation and fibrosis in a rat model of chronic

cyclosporine-induced nephropathy. Am J Physiol Renal Physiol2004; 286: F46–F57

45. Vieira JM, Jr, Mantovani E, Rodrigues LT et al. Simvastatinattenuates renal inflammation, tubular transdifferentiation and

interstitial fibrosis in rats with unilateral ureteral obstruction.

Nephrol Dial Transplant 2005; 20: 1582–159146. Agarwal R. Effects of statins on renal function. Am J Cardiol

2006; 97: 748–5547. Fujihara CK, De Lourdes NI, Malheiros, AGR, de Oliveira IB,

Zatz R. Combined mycophenolate mofetil and losartan therapy

arrests established injury in the remnant kidney. J Am Soc

Nephrol 2000; 11: 283–29048. Goncalves RG, Biato MA, Colosimo RD et al. Effects of

mycophenolate mofetil and lisinopril on collagen deposition in

unilateral ureteral obstruction in rats. Am J Nephrol 2004; 24:

527–53649. Kramer S, Loof T, Martini S et al. Mycophenolate mofetil

slows progression in anti-thy1-induced chronic renal fibrosis but

is not additive to a high dose of enalapril. Am J Physiol Renal

Physiol 2005; 289: F359–F36850. Romero F, Rodriguez-Iturbe B, Parra G, Gonzalez L, Herrera-

Acosta J, Tapia E. Mycophenolate mofetil prevents theprogressive renal failure induced by 5/6 renal ablation in rats.Kidney Int 1999; 55: 945–955

51. Gwinner W, Mengel M, Franz I et al. Effect of mycophenolatemofetil (mmf) on tubulointerstitial fibrosis and tubular atrophyin renal allograft recipients studied by protocol biopsies.Transplantation 2006; 82: 663–664

52. Mota A, Arias M, Taskinen EI et al. Sirolimus-based therapyfollowing early cyclosporine withdrawal provides significantlyimproved renal histology and function at 3 years. Am JTransplant 2004; 4: 953–961

53. Wu MJ, Wen MC, Chiu YT et al. Rapamycin attenuatesunilateral ureteral obstruction-induced renal fibrosis. Kidney Int2006; 69: 2029–2036

54. Peters H, Martini S, Wang Y et al. Selective lymphocyteinhibition by FTY720 slows the progressive course of chronicanti-thy 1 glomerulosclerosis. Kidney Int 2004; 66: 1434–1443

55. Muller DN, Shagdarsuren E, Park JK et al. Immunosuppressivetreatment protects against angiotensin II-induced renal damage.Am J Pathol 2002; 161: 1679–1693

56. Suzuki K, Kanabayashi T, Nakayama H, Doi K. Effects oftacrolimus and dexamethasone on tubulointerstitial fibrosis inmercuric chloride treated Brown Norway rats. Exp ToxicolPathol 2003; 55: 197–207

57. Catania JM, Chen G, Parrish AR. Role of matrix metallopro-teinases in renal pathophysiologies. Am J Physiol Renal Physiol2007; 292: F905–F911

58. Aoyama T, Yamamoto S, Kanematsu A, Ogawa O, Tabata Y.Local delivery of matrix metalloproteinase gene prevents theonset of renal sclerosis in streptozotocin-induced diabetic mice.Tissue Eng 2003; 9: 1289–1299

59. Cheng S, Pollock AS, Mahimkar R, Olson JL, Lovett DH.Matrix metalloproteinase 2 and basement membrane integrity: aunifying mechanism for progressive renal injury. FASEB J 2006;20: 1898–1900

60. Nishida M, Okumura Y, Ozawa S, Shiraishi I, Itoi T,Hamaoka K. MMP-2 inhibition reduces renal macrophageinfiltration with increased fibrosis in UUO. Biochem Biophys ResCommun 2007; 354: 133–139

61. Kim H, Oda T, Lopez-Guisa J et al. TIMP-1 deficiency does notattenuate interstitial fibrosis in obstructive nephropathy. J AmSoc Nephrol 2001; 12: 736–748

62. Eddy AA, Kim H, Lopez-Guisa J, Oda T, Soloway PD.Interstitial fibrosis in mice with overload proteinuria:deficiency of TIMP-1 is not protective. Kidney Int 2000; 58:618–628

63. Zhang X, Chen X, Hong Q et al. TIMP-1 promotes age-relatedrenal fibrosis through upregulating ICAM-1 in human TIMP-1transgenic mice. J Gerontol A Biol Sci Med Sci 2006; 61:1130–1143

64. Melenhorst WB, van den Heuvel MC, Timmer A et al.ADAM19 expression in human nephrogenesis and renal disease:associations with clinical and structural deterioration. Kidney Int2006; 70: 1269–1278

65. Mittaz L, Ricardo S, Martinez G et al. Neonatal calycealdilation and renal fibrosis resulting from loss of Adamts-1 inmouse kidney is due to a developmental dysgenesis. Nephrol DialTransplant 2005; 20: 419–423

66. Eddy AA, Fogo AB. Plasminogen activator inhibitor-1 inchronic kidney disease: evidence and mechanisms of action.J Am Soc Nephrol 2006; 17: 2999–3012

67. Huang Y, Haraguchi M, Lawrence DA, Border WA, Yu L,Noble NA. A mutant, noninhibitory plasminogen activatorinhibitor type 1 decreases matrix accumulation in experimentalglomerulonephritis. J Clin Invest 2003; 112: 379–388

68. Kitching AR, Kong YZ, Huang XR et al. Plasminogen activatorinhibitor-1 is a significant determinant of renal injury in

Treatment targets in renal fibrosis 3401

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 12: View - Nephrology Dialysis Transplantation

experimental crescentic glomerulonephritis. J Am Soc Nephrol2003; 14: 1487–1495

69. Krag S, Danielsen CC, Carmeliet P, Nyengaard J, Wogensen L.Plasminogen activator inhibitor-1 gene deficiency attenuatesTGF-beta1-induced kidney disease. Kidney Int 2005; 68:2651–2666

70. Yang J, Shultz RW, Mars WM et al. Disruption of tissue-typeplasminogen activator gene in mice reduces renal interstitialfibrosis in obstructive nephropathy. J Clin Invest 2002; 110:1525–1538

71. Haraguchi M, Border WA, Huang Y, Noble NA. t-PA promotesglomerular plasmin generation and matrix degradation inexperimental glomerulonephritis. Kidney Int 2001; 59: 2146–2155

72. Edgtton KL, Gow RM, Kelly DJ, Carmeliet P, Kitching AR.Plasmin is not protective in experimental renal interstitialfibrosis. Kidney Int 2004; 66: 68–76

73. Zhang G, Kernan KA, Collins SJ et al. Plasmin(ogen) promotesrenal interstitial fibrosis by promoting epithelial-to-mesenchy-mal transition: role of plasmin-activated signals. J Am SocNephrol 2007; 18: 846–859

74. Kitching AR, Holdsworth SR, Ploplis VA et al. Plasminogenand plasminogen activators protect against renal injury increscentic glomerulonephritis. J Exp Med 1997; 185: 963–968

75. Hertig A, Berrou J, Allory Y et al. Type 1 plasminogen activatorinhibitor deficiency aggravates the course of experimentalglomerulonephritis through overactivation of transforminggrowth factor beta. FASEB J 2003; 17: 1904–1906

76. Johnson TS, El-Koraie AF, Skill NJ et al. Tissue transglutami-nase and the progression of human renal scarring. J Am SocNephrol 2003; 14: 2052–2062

77. Skill NJ, Johnson TS, Coutts IG et al. Inhibition of transglu-taminase activity reduces extracellular matrix accumulationinduced by high glucose levels in proximal tubular epithelialcells. J Biol Chem 2004; 279: 47754–47762

78. Sebekova K, Dammrich J, Fierlbeck W, Krivosikova Z,Paczek L, Heidland A. Effect of chronic therapy withproteolytic enzymes on hypertension-induced renal injury inthe rat model of Goldblatt hypertension. Am J Nephrol 1998; 18:570–576

79. Sebekova K, Paczek L, Dammrich J et al. Effects of proteasetherapy in the remnant kidney model of progressive renal failure.Miner Electrolyte Metab 1997; 23: 291–295

80. Danielson LA, Welford A, Harris A. Relaxin improves renalfunction and histology in aging Munich Wistar rats. J Am SocNephrol 2006; 17: 1325–1333

81. Hewitson TD, Mookerjee I, Masterson R et al. Endogenousrelaxin is a naturally occurring modulator of experimental renaltubulointerstitial fibrosis. Endocrinology 2007; 148: 660–669

82. Lekgabe ED, Kiriazis H, Zhao C et al. Relaxin reverses cardiacand renal fibrosis in spontaneously hypertensive rats.Hypertension 2005; 46: 412–418

83. Samuel CS, Hewitson TD. Relaxin in cardiovascular and renaldisease. Kidney Int 2006; 69: 1498–1502

84. Seibold JR. Relaxins: lessons and limitations. Curr RheumatolRep 2002; 4: 275–276

85. Leh S, Vaagnes O, Margolin SB, Iversen BM, Forslund T.Pirfenidone and candesartan ameliorate morphological damagein mild chronic anti-GBM nephritis in rats. Nephrol DialTransplant 2005; 20: 71–82

86. Negri AL. Prevention of progressive fibrosis in chronic renaldiseases: antifibrotic agents. J Nephrol 2004; 17: 496–503

87. Chen X, Moeckel G, Morrow JD et al. Lack of integrinalpha1beta1 leads to severe glomerulosclerosis after glomerularinjury. Am J Pathol 2004; 165: 617–630

88. Cosgrove D, Rodgers K, Meehan D et al. Integrin alpha1beta1and transforming growth factor-beta1 play distinct roles inalport glomerular pathogenesis and serve as dual targets formetabolic therapy. Am J Pathol 2000; 157: 1649–1659

89. Cook HT, Khan SB, Allen A et al. Treatment with an antibodyto VLA-1 integrin reduces glomerular and tubulointerstitial

scarring in a rat model of crescentic glomerulonephritis. Am JPathol 2002; 161: 1265–1272

90. Kagami S, Urushihara M, Kondo S et al. Effects of anti-alpha1integrin subunit antibody on anti-Thy-1 glomerulonephritis.Lab Invest 2002; 82: 1219–27

91. Ma LJ, Yang H, Gaspert A et al. Transforming growth factor-beta-dependent and -independent pathways of induction of

tubulointerstitial fibrosis in beta6(-/-) mice. Am J Pathol 2003;163: 1261–1273

92. Hahm K, Lukashev ME, Luo Y et al. {alpha}v{beta}6 Integrinregulates renal fibrosis and inflammation in alport mouse.Am J Pathol 2007; 170: 110–125

93. Hartner A, Cordasic N, Klanke B, Muller U, Sterzel RB,Hilgers KF. The alpha8 integrin chain affords mechanical

stability to the glomerular capillary tuft in hypertensiveglomerular disease. Am J Pathol 2002; 160: 861–867

94. Blattner SM, Kretzler M. Integrin-linked kinase in renaldisease: connecting cell-matrix interaction to the cytoskeleton.Curr Opin Nephrol Hypertens 2005; 14: 404–410

95. Li Y, Yang J, Dai C, Wu C, Liu Y. Role for integrin-linked

kinase in mediating tubular epithelial to mesenchymal transi-tion and renal interstitial fibrogenesis. J Clin Invest 2003; 112:503–516

96. Couser WG, Ochi RF, Baker PJ, Schulze M, Campbell C,Johnson RJ. C6 depletion reduces proteinuria in experimentalnephropathy induced by a nonglomerular antigen. J Am Soc

Nephrol 1991; 2: 894–90197. Morita Y, Nomura A, Yuzawa Y et al. The role of complement

in the pathogenesis of tubulointerstitial lesions in rat mesangialproliferative glomerulonephritis. J Am Soc Nephrol 1997; 8:1363–1372

98. Nomura A, Morita Y, Maruyama S et al. Role of complement

in acute tubulointerstitial injury of rats with aminonucleosidenephrosis. Am J Pathol 1997; 151: 539–547

99. Rangan GK, Pippin JW, Couser WG. C5b-9 regulatesperitubular myofibroblast accumulation in experimental focalsegmental glomerulosclerosis. Kidney Int 2004; 66: 1838–1848

100. Rangan GK, Pippin JW, Coombes JD, Couser WG. C5b-9does not mediate chronic tubulointerstitial disease in the

absence of proteinuria. Kidney Int 2005; 67: 492–503101. Bao L, Wang Y, Chang A et al. Unrestricted c3 activation

occurs in crry-deficient kidneys and rapidly leads to chronicrenal failure. J Am Soc Nephrol 2007; 18: 811–822

102. Hori Y, Yamada K, Hanafusa N et al. Crry, a complementregulatory protein, modulates renal interstitial disease inducedby proteinuria. Kidney Int 1999; 56: 2096–2106

103. He C, Imai M, Song H, Quigg RJ, Tomlinson S. Complement

inhibitors targeted to the proximal tubule prevent injury inexperimental nephrotic syndrome and demonstrate a key rolefor C5b-9. J Immunol 2005; 174: 5750–5757

104. Bao L, Zhou J, Holers VM, Quigg RJ. Excessive matrixaccumulation in the kidneys of MRL/lpr lupus mice is

dependent on complement activation. J Am Soc Nephrol2003; 14: 2516–2525

105. Welch TR, Frenzke M, Witte D, Davis AE. C5a is important inthe tubulointerstitial component of experimental immune

complex glomerulonephritis. Clin Exp Immunol 2002; 130:

43–48106. Boor P, Konieczny A, Villa L et al. Complement c5 mediates

experimental tubulointerstitial fibrosis. J Am Soc Nephrol 2007;

18: 1508–1515107. Bao L, Osawe I, Puri T, Lambris JD, Haas M, Quigg RJ. C5a

promotes development of experimental lupus nephritis which

can be blocked with a specific receptor antagonist. Eur J

Immunol 2005; 35: 2496–2506108. Wenderfer SE, Ke B, Hollmann TJ, Wetsel RA, Lan HY,

Braun MC. C5a receptor deficiency attenuates T cell function

and renal disease in MRLlpr mice. J Am Soc Nephrol 2005; 16:

3572–3582

3402 P. Boor et al.

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 13: View - Nephrology Dialysis Transplantation

109. Fan JM, Huang XR, Ng YY et al. Interleukin-1 inducestubular epithelial-myofibroblast transdifferentiation through atransforming growth factor-beta1-dependent mechanismin vitro. Am J Kidney Dis 2001; 37: 820–831

110. Lan HY, Nikolic-Paterson DJ, Mu W, Vannice JL, Atkins RC.Interleukin-1 receptor antagonist halts the progression ofestablished crescentic glomerulonephritis in the rat. KidneyInt 1995; 47: 1303–1309

111. Taal MW, Zandi-Nejad K, Weening B et al. Proinflammatorygene expression and macrophage recruitment in the ratremnant kidney. Kidney Int 2000; 58: 1664–1676

112. Vesey DA, Cheung CW, Cuttle L, Endre ZA, Gobe G,Johnson DW. Interleukin-1beta induces human proximaltubule cell injury, alpha-smooth muscle actin expression andfibronectin production. Kidney Int 2002; 62: 31–40

113. Yamagishi H, Yokoo T, Imasawa T, Mitarai T, Kawamura T,Utsunomiya Y. Genetically modified bone marrow-derivedvehicle cells site specifically deliver an anti-inflammatorycytokine to inflamed interstitium of obstructive nephropathy.J Immunol 2001; 166: 609–616

114. Svensson M, Irjala H, Alm P, Holmqvist B, Lundstedt AC,Svanborg C. Natural history of renal scarring in susceptiblemIL-8Rh-/- mice. Kidney Int 2005; 67: 103–110

115. Mu W, Ouyang X, Agarwal A et al. IL-10 suppresseschemokines, inflammation, and fibrosis in a model of chronicrenal disease. J Am Soc Nephrol 2005; 16: 3651–3660

116. Asadullah K, Sterry W, Volk HD. Interleukin-10 therapy–review of a new approach. Pharmacol Rev 2003; 55: 241–269

117. Cook HT, Singh SJ, Wembridge DE, Smith J, Tam FW,Pusey CD. Interleukin-4 ameliorates crescentic glomerulone-phritis in Wistar Kyoto rats. Kidney Int 1999; 55: 1319–1326

118. Strutz F, Heeg M, Kochsiek T, Siemers G, Zeisberg M,Muller GA. Effects of pentoxifylline, pentifylline and gamma-interferon on proliferation, differentiation, and matrixsynthesis of human renal fibroblasts. Nephrol Dial Transplant2000; 15: 1535–1546

119. Oldroyd SD, Thomas GL, Gabbiani G, El Nahas AM.Interferon-gamma inhibits experimental renal fibrosis. KidneyInt 1999; 56: 2116–2127

120. Johnson RJ, Lombardi D, Eng E et al. Modulation ofexperimental mesangial proliferative nephritis by interferon-gamma. Kidney Int 1995; 47: 62–69

121. Dogukan A, Akpolat N, Celiker H, Ilhan N, HalilBahcecioglu I, Gunal AI. Protective effect of interferon-alphaon carbon tetrachloride-induced nephrotoxicity. J Nephrol2003; 16: 81–84

122. Guo G, Morrissey J, McCracken R, Tolley T, Liapis H,Klahr S. Contributions of angiotensin II and tumor necrosisfactor-alpha to the development of renal fibrosis. Am J PhysiolRenal Physiol 2001; 280: F777–F785

123. Khan SB, Cook HT, Bhangal G, Smith J, Tam FW, Pusey CD.Antibody blockade of TNF-alpha reduces inflammation andscarring in experimental crescentic glomerulonephritis.Kidney Int 2005; 67: 1812–1820

124. Meldrum KK, Misseri R, Metcalfe P, Dinarello CA, Hile KL,Meldrum DR. TNF-{alpha} neutralization amelioratesobstruction-induced renal fibrosis and dysfunction. Am JPhysiol Regul Integr Comp Physiol 2007; 292: R1456–R1464

125. Bongartz T, Sutton AJ, Sweeting MJ, Buchan I, Matteson EL,Montori V. Anti-TNF antibody therapy in rheumatoid arthritisand the risk of serious infections and malignancies: systematicreview and meta-analysis of rare harmful effects in randomizedcontrolled trials. JAMA 2006; 295: 2275–2285

126. Mann DL, McMurray JJ, Packer M et al. Targeted antic-ytokine therapy in patients with chronic heart failure: results ofthe Randomized Etanercept Worldwide Evaluation(RENEWAL). Circulation 2004; 109: 1594–1602

127. Kitagawa K, Wada T, Furuichi K et al. Blockade of CCR2ameliorates progressive fibrosis in kidney. Am J Pathol 2004;165: 237–246

128. Lloyd CM, Minto AW, Dorf ME et al. RANTES andmonocyte chemoattractant protein-1 (MCP-1) play an impor-tant role in the inflammatory phase of crescentic nephritis, butonly MCP-1 is involved in crescent formation and interstitialfibrosis. J Exp Med 1997; 185: 1371–1380

129. Okada H, Moriwaki K, Kalluri R et al. Inhibition of monocytechemoattractant protein-1 expression in tubular epitheliumattenuates tubulointerstitial alteration in rat Goodpasturesyndrome. Kidney Int 2000; 57: 927–936

130. Tesch GH, Maifert S, Schwarting A, Rollins BJ, Kelley VR.Monocyte chemoattractant protein 1-dependent leukocyticinfiltrates are responsible for autoimmune disease in MRL-Fas(lpr) mice. J Exp Med 1999; 190: 1813–1824

131. Tesch GH, Schwarting A, Kinoshita K, Lan HY, Rollins BJ,Kelley VR. Monocyte chemoattractant protein-1 promotesmacrophage-mediated tubular injury, but not glomerularinjury, in nephrotoxic serum nephritis. J Clin Invest 1999;103: 73–80

132. Vielhauer V, Anders HJ, Mack M et al. Obstructive nephro-pathy in the mouse: progressive fibrosis correlates withtubulointerstitial chemokine expression and accumulation ofCC chemokine receptor 2- and 5-positive leukocytes. J Am SocNephrol 2001; 12: 1173–1187

133. Wada T, Furuichi K, Sakai N et al. Gene therapy via blockadeof monocyte chemoattractant protein-1 for renal fibrosis. J AmSoc Nephrol 2004; 15: 940–948

134. Anders HJ, Vielhauer V, Frink M et al. A chemokine receptorCCR-1 antagonist reduces renal fibrosis after unilateral ureterligation. J Clin Invest 2002; 109: 251–259

135. Eis V, Luckow B, Vielhauer V et al. Chemokine receptor CCR1but not CCR5 mediates leukocyte recruitment and subsequentrenal fibrosis after unilateral ureteral obstruction. J Am SocNephrol 2004; 15: 337–347

136. Ninichuk V, Gross O, Reichel C et al. Delayed chemokinereceptor 1 blockade prolongs survival in collagen 4A3-deficientmice with Alport disease. J Am Soc Nephrol 2005; 16: 977–985

137. Vielhauer V, Berning E, Eis V et al. CCR1 blockade reducesinterstitial inflammation and fibrosis in mice with glomerulo-sclerosis and nephrotic syndrome. Kidney Int 2004; 66:2264–2278

138. Lenda DM, Kikawada E, Stanley ER, Kelley VR. Reducedmacrophage recruitment, proliferation, and activation incolony-stimulating factor-1-deficient mice results in decreasedtubular apoptosis during renal inflammation. J Immunol 2003;170: 3254–3262

139. Ophascharoensuk V, Giachelli CM, Gordon K et al.Obstructive uropathy in the mouse: role of osteopontin ininterstitial fibrosis and apoptosis. Kidney Int 1999; 56: 571–580

140. Panzer U, Thaiss F, Zahner G et al.Monocyte chemoattractantprotein-1 and osteopontin differentially regulate monocytesrecruitment in experimental glomerulonephritis. Kidney Int2001; 59: 1762–1769

141. Yoo KH, Thornhill BA, Forbes MS et al. Osteopontinregulates renal apoptosis and interstitial fibrosis in neonatalchronic unilateral ureteral obstruction. Kidney Int 2006; 70:1735–1741

142. Furuichi K, Gao JL, Murphy PM. Chemokine receptorCX3CR1 regulates renal interstitial fibrosis after ischemia-reperfusion injury. Am J Pathol 2006; 169: 372–387

143. Sakai N, Wada T, Yokoyama H et al. Secondary lymphoidtissue chemokine (SLC/CCL21)/CCR7 signaling regulatesfibrocytes in renal fibrosis. Proc Natl Acad Sci USA 2006;103: 14098–14103

144. Topham PS, Csizmadia V, Soler D et al. Lack of chemokinereceptor CCR1 enhances Th1 responses and glomerular injuryduring nephrotoxic nephritis. J Clin Invest 1999; 104:1549–1557

145. Bird JE, Giancarli MR, Kurihara T et al. Increased severity ofglomerulonephritis in C-C chemokine receptor 2 knockoutmice. Kidney Int 2000; 57: 129–136

Treatment targets in renal fibrosis 3403

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 14: View - Nephrology Dialysis Transplantation

146. Barber DF, Bartolome A, Hernandez C et al. PI3Kgammainhibition blocks glomerulonephritis and extends lifespan in amouse model of systemic lupus. Nat Med 2005; 11: 933–935

147. Bottinger EP, Bitzer M. TGF-beta signaling in renal disease.J Am Soc Nephrol 2002; 13: 2600–2610

148. Shull MM, Ormsby I, Kier AB et al. Targeted disruption of themouse transforming growth factor-beta 1 gene results inmultifocal inflammatory disease. Nature 1992; 359: 693–699

149. Yu L, Border WA, Huang Y, Noble NA. TGF-beta isoforms inrenal fibrogenesis. Kidney Int 2003; 64: 844–856

150. Terada Y, Hanada S, Nakao A, Kuwahara M, Sasaki S,Marumo F. Gene transfer of Smad7 using electroporation ofadenovirus prevents renal fibrosis in post-obstructed kidney.Kidney Int 2002; 61: 94–98

151. Lan HY, Mu W, Tomita N et al. Inhibition of renal fibrosis bygene transfer of inducible Smad7 using ultrasound-microbubblesystem in rat UUO model. J Am Soc Nephrol 2003; 14:1535–1548

152. Hou CC, Wang W, Huang XR et al. Ultrasound-microbubble-mediated gene transfer of inducible Smad7 blocks transforminggrowth factor-beta signaling and fibrosis in rat remnant kidney.Am J Pathol 2005; 166: 761–71

153. Inazaki K, Kanamaru Y, Kojima Y et al. Smad3 deficiencyattenuates renal fibrosis, inflammation,and apoptosis afterunilateral ureteral obstruction. Kidney Int 2004; 66: 597–604

154. Sato M, Muragaki Y, Saika S, Roberts AB, Ooshima A.Targeted disruption of TGF-beta1/Smad3 signaling protectsagainst renal tubulointerstitial fibrosis induced by unilateralureteral obstruction. J Clin Invest 2003; 112: 1486–1494

155. Nagler A, Katz A, Aingorn H et al. Inhibition of glomerularmesangial cell proliferation and extracellular matrix depositionby halofuginone. Kidney Int 1997; 52: 1561–1569

156. Boutet A, De Frutos CA, Maxwell PH, Mayol MJ, Romero J,Nieto MA. Snail activation disrupts tissue homeostasis andinduces fibrosis in the adult kidney. Embo J 2006; 25:5603–5613

157. Fukasawa H, Yamamoto T, Togawa A et al. Ubiquitin-dependent degradation of SnoN and Ski is increased in renalfibrosis induced by obstructive injury. Kidney Int 2006; 69:1733–1740

158. Yang J, Zhang X, Li Y, Liu Y. Downregulation of Smadtranscriptional corepressors SnoN and Ski in the fibrotickidney: an amplification mechanism for TGF-beta1 signaling.J Am Soc Nephrol 2003; 14: 3167–3177

159. Moon JA, Kim HT, Cho IS, Sheen YY, Kim DK. IN-1130,a novel transforming growth factor-beta type I receptor kinase(ALK5) inhibitor, suppresses renal fibrosis in obstructivenephropathy. Kidney Int 2006; 70: 1234–1243

160. Grygielko ET, Martin WM, Tweed C et al. Inhibition of genemarkers of fibrosis with a novel inhibitor of transforminggrowth factor-beta type I receptor kinase in puromycin-inducednephritis. J Pharmacol Exp Ther 2005; 313: 943–951

161. Li J, Campanale NV, Liang RJ, Deane JA, Bertram JF,Ricardo SD. Inhibition of p38 mitogen-activated proteinkinase and transforming growth factor-beta1/Smad signalingpathways modulates the development of fibrosis in adriamycin-induced nephropathy. Am J Pathol 2006; 169: 1527–1540

162. Prakash J, Sandovici M, Saluja V et al. Intracellular deliveryof the p38 mitogen-activated protein kinase inhibitorSB202190 [4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)1H-imidazole] in renal tubular cells: a novel strategy to treatrenal fibrosis. J Pharmacol Exp Ther 2006; 319: 8–19

163. Stambe C, Atkins RC, Tesch GH, Masaki T, Schreiner GF,Nikolic-Paterson DJ. The role of p38alpha mitogen-activatedprotein kinase activation in renal fibrosis. J Am Soc Nephrol2004; 15: 370–379

164. Schaefer L, Macakova K, Raslik I et al. Absence of decorinadversely influences tubulointerstitial fibrosis of the obstructedkidney by enhanced apoptosis and increased inflammatoryreaction. Am J Pathol 2002; 160: 1181–1191

165. Isaka Y, Brees DK, Ikegaya K et al. Gene therapy by skeletalmuscle expression of decorin prevents fibrotic disease in ratkidney. Nat Med 1996; 2: 418–423

166. Zeisberg M, Hanai J, Sugimoto H et al. BMP-7 counteractsTGF-beta1-induced epithelial-to-mesenchymal transition andreverses chronic renal injury. Nat Med 2003; 9: 964–968

167. Zeisberg M, Bottiglio C, Kumar N et al. Bone morphogenicprotein-7 inhibits progression of chronic renal fibrosis asso-ciated with two genetic mouse models. Am J Physiol RenalPhysiol 2003; 285: F1060–F1067

168. Zeisberg M, Shah AA, Kalluri R. Bone morphogenic protein-7induces mesenchymal to epithelial transition in adult renalfibroblasts and facilitates regeneration of injured kidney. J BiolChem 2005; 280: 8094–8100

169. Hruska KA, Guo G, Wozniak M et al. Osteogenic protein-1prevents renal fibrogenesis associated with ureteral obstruction.

Am J Physiol Renal Physiol 2000; 279: F130–F143170. Wang S, Chen Q, Simon TC et al. Bone morphogenic protein-7

(BMP-7), a novel therapy for diabetic nephropathy. Kidney Int2003; 63: 2037–2049

171. Li T, Surendran K, Zawaideh MA, Mathew S, Hruska KA.Bone morphogenetic protein 7: a novel treatment for chronicrenal and bone disease. Curr Opin Nephrol Hypertens 2004; 13:417–422

172. Zeisberg M. Bone morphogenic protein-7 and the kidney:current concepts and open questions. Nephrol Dial Transplant2006; 21: 568–573

173. Ikeda Y, Jung YO, Kim H et al. Exogenous bone morphoge-netic protein-7 fails to attenuate renal fibrosis in rats withoverload proteinuria. Nephron Exp Nephrol 2004; 97:e123–e135

174. Lin J, Patel SR, Cheng X et al. Kielin/chordin-like protein, anovel enhancer of BMP signaling, attenuates renal fibroticdisease. Nat Med 2005; 11: 387–393

175. Yanagita M. Modulator of bone morphogenetic proteinactivity in the progression of kidney diseases. Kidney Int

2006; 70: 989–993176. Herrero-Fresneda I, Torras J, Franquesa M et al. HGF gene

therapy attenuates renal allograft scarring by preventing theprofibrotic inflammatory-induced mechanisms. Kidney Int2006; 70: 265–274

177. Liu Y. Hepatocyte growth factor in kidney fibrosis: therapeuticpotential and mechanisms of action. Am J Physiol RenalPhysiol 2004; 287: F7–F16

178. Mizuno S, Kurosawa T, Matsumoto K, Mizuno-Horikawa Y,Okamoto M, Nakamura T. Hepatocyte growth factor preventsrenal fibrosis and dysfunction in a mouse model of chronicrenal disease. J Clin Invest 1998; 101: 1827–1834

179. Mizuno S, Matsumoto K, Nakamura T. Hepatocyte growthfactor suppresses interstitial fibrosis in a mouse model ofobstructive nephropathy. Kidney Int 2001; 59: 1304–1314

180. Takayama H, LaRochelle WJ, Sabnis SG, Otsuka T,Merlino G. Renal tubular hyperplasia, polycystic disease, and

glomerulosclerosis in transgenic mice overexpressing hepato-

cyte growth factor/scatter factor. Lab Invest 1997; 77: 131–138181. Wang SN, LaPage J, Hirschberg R. Role of glomerular

ultrafiltration of growth factors in progressive interstitial

fibrosis in diabetic nephropathy. Kidney Int 2000; 57:

1002–1014182. Jiang WG, Martin TA, Parr C, Davies G, Matsumoto K,

Nakamura T. Hepatocyte growth factor, its receptor, and their

potential value in cancer therapies. Crit Rev Oncol Hematol

2005; 53: 35–69183. Abdel WN, Mason RM. Connective tissue growth factor and

renal diseases: some answers, more questions. Curr Opin

Nephrol Hypertens 2004; 13: 53–58184. van Nieuwenhoven FA, Jensen LJ, Flyvbjerg A,

Goldschmeding R. Imbalance of growth factor signalling in

diabetic kidney disease: is connective tissue growth factor

3404 P. Boor et al.

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 15: View - Nephrology Dialysis Transplantation

(CTGF, CCN2) the perfect intervention point? Nephrol Dial

Transplant 2005; 20: 6–10185. Burns WC, Twigg SM, Forbes JM et al. Connective tissue

growth factor plays an important role in advanced glycationend product-induced tubular epithelial-to-mesenchymal transi-tion: implications for diabetic renal disease. J Am Soc Nephrol

2006; 17: 2484–2494186. Okada H, Kikuta T, Kobayashi T et al. Connective tissue

growth factor expressed in tubular epithelium plays a pivotalrole in renal fibrogenesis. J Am Soc Nephrol 2005; 16: 133–143

187. Yokoi H, Mukoyama M, Nagae T et al. Reduction in

connective tissue growth factor by antisense treatment amelio-rates renal tubulointerstitial fibrosis. J Am Soc Nephrol 2004;15: 1430–1440

188. Zhou G, Li C, Cai L. Advanced glycation end-products

induce connective tissue growth factor-mediated renalfibrosis predominantly through transforming growthfactor beta-independent pathway. Am J Pathol 2004; 165:

2033–2043189. Kang DH, Johnson RJ. Vascular endothelial growth factor:

a new player in the pathogenesis of renal fibrosis. Curr OpinNephrol Hypertens 2003; 12: 43–49

190. Kang DH, Hughes J, Mazzali M, Schreiner GF, Johnson RJ.Impaired angiogenesis in the remnant kidney model: II.

Vascular endothelial growth factor administration reducesrenal fibrosis and stabilizes renal function. J Am Soc Nephrol2001; 12: 1448–1457

191. Kang DH, Kim YG, Andoh TF et al. Post-cyclosporine-

mediated hypertension and nephropathy: amelioration byvascular endothelial growth factor. Am J Physiol RenalPhysiol 2001; 280: F727–F736

192. Hara A, Wada T, Furuichi K et al. Blockade of VEGF

accelerates proteinuria, via decrease in nephrin expressionin rat crescentic glomerulonephritis. Kidney Int 2006; 69:1986–1995

193. Ostendorf T, Kunter U, Eitner F et al. VEGF(165)

mediates glomerular endothelial repair. J Clin Invest 1999;104: 913–923

194. de Vriese AS, Tilton RG, Elger M, Stephan CC, Kriz W,Lameire NH. Antibodies against vascular endothelial growth

factor improve early renal dysfunction in experimental dia-betes. J Am Soc Nephrol 2001; 12: 993–1000

195. Schrijvers BF, Flyvbjerg A, Tilton RG, Rasch R, Lameire NH,De Vriese AS. Pathophysiological role of vascular endothelial

growth factor in the remnant kidney. Nephron Exp Nephrol2005; 101: e9–e15

196. Schrijvers BF, Rasch R, Tilton RG, Flyvbjerg A. High protein-induced glomerular hypertrophy is vascular endothelial growth

factor-dependent. Kidney Int 2002; 61: 1600–1604197. Flyvbjerg A, Dagnaes-Hansen F, De Vriese AS, Schrijvers BF,

Tilton RG, Rasch R. Amelioration of long-term renal changesin obese type 2 diabetic mice by a neutralizing vascular

endothelial growth factor antibody. Diabetes 2002; 51:3090–3094

198. Herbst RS. Toxicities of antiangiogenic therapy in non-small-cell lung cancer. Clin Lung Cancer 2006; 8 [Suppl 1]:

S23–S30199. Floege J, Eitner F, Alpers CE. A new look at platelet-derived

growth factor and renal disease. J Am Soc Nephrol 2007 in

press200. Ostendorf T, Kunter U, Grone HJ et al. Specific antagonism of

PDGF prevents renal scarring in experimental glomerulone-

phritis. J Am Soc Nephrol 2001; 12: 909–918201. Tang WW, Ulich TR, Lacey DL et al. Platelet-derived growth

factor-BB induces renal tubulointerstitial myofibroblast for-mation and tubulointerstitial fibrosis. Am J Pathol 1996; 148:1169–1180

202. Taneda S, Hudkins KL, Topouzis S et al. Obstructive uropathyin mice and humans: potential role for PDGF-D in the

progression of tubulointerstitial injury. J Am Soc Nephrol

2003; 14: 2544–2555203. Ostendorf T, Rong S, Boor P et al. Antagonism of PDGF-D

by human antibody CR002 prevents renal scarring in experi-mental glomerulonephritis. J Am Soc Nephrol 2006; 17:1054–1062

204. Boor P, Konieczny A, Villa L et al. PDGF-D inhibition by

CR002 ameliorates tubulointerstitial fibrosis following experi-mental glomerulonephritis. Nephrol Dial Transplant 2007; 22:1323–1331

205. Eitner F, Ostendorf T, Kretzler M et al. PDGF-C expression in

the developing and normal adult human kidney and inglomerular diseases. J Am Soc Nephrol 2003; 14: 1145–1153

206. Eitner F, Ostendorf T, Van Roeyen C et al. Expression of anovel PDGF isoform, PDGF-C, in normal and diseased rat

kidney. J Am Soc Nephrol 2002; 13: 910–917207. Wang S, Wilkes MC, Leof EB, Hirschberg R. Imatinib

mesylate blocks a non-Smad TGF-beta pathway and reducesrenal fibrogenesis in vivo. FASEB J 2005; 19: 1–11

208. Lassila M, Jandeleit-Dahm K, Seah KK et al. Imatinibattenuates diabetic nephropathy in apolipoprotein E-knockout

mice. J Am Soc Nephrol 2005; 16: 363–373209. Savikko J, Taskinen E, Von Willebrand E. Chronic allograft

nephropathy is prevented by inhibition of platelet-derivedgrowth factor receptor: tyrosine kinase inhibitors as a potential

therapy. Transplantation 2003; 75: 1147–1153210. Kerkela R, Grazette L, Yacobi R et al. Cardiotoxicity of the

cancer therapeutic agent imatinib mesylate. Nat Med 2006; 12:908–916

211. Berman E, Nicolaides M, Maki RG et al. Altered bone and

mineral metabolism in patients receiving imatinib mesylate.N Engl J Med 2006; 354: 2006–2013

212. Floege J, Kriz W, Schulze M et al. Basic fibroblast growthfactor augments podocyte injury and induces glomerulosclero-

sis in rats with experimental membranous nephropathy. J ClinInvest 1995; 96: 2809–2819

213. Strutz F, Zeisberg M, Hemmerlein B et al. Basic fibroblastgrowth factor expression is increased in human renal fibrogen-

esis and may mediate autocrine fibroblast proliferation. KidneyInt 2000; 57: 1521–1538

214. Strutz F, Zeisberg M, Ziyadeh FN et al. Role of basic fibroblastgrowth factor-2 in epithelial-mesenchymal transformation.

Kidney Int 2002; 61: 1714–1728215. Rossini M, Cheunsuchon B, Donnert E et al.

Immunolocalization of fibroblast growth factor-1 (FGF-1),its receptor (FGFR-1), and fibroblast-specific protein-1

(FSP-1) in inflammatory renal disease. Kidney Int 2005; 68:2621–2628

216. Francois H, Placier S, Flamant M et al. Prevention of renalvascular and glomerular fibrosis by epidermal growth factorreceptor inhibition. FASEB J 2004; 18: 926–928

217. Terzi F, Burtin M, Hekmati M et al. Targeted expression of a

dominant-negative EGF-R in the kidney reduces tubulo-interstitial lesions after renal injury. J Clin Invest 2000; 106:225–234

218. Lautrette A, Li S, Alili R et al. Angiotensin II and EGF

receptor cross-talk in chronic kidney diseases: a new therapeu-tic approach. Nat Med 2005; 11: 867–874

219. Morrissey JJ, Ishidoya S, McCracken R, Klahr S. Nitricoxide generation ameliorates the tubulointerstitial fibrosis

of obstructive nephropathy. J Am Soc Nephrol 1996; 7:

2202–2212220. Peters H, Daig U, Martini S et al. NO mediates antifibrotic

actions of L-arginine supplementation following induction of

anti-thy1 glomerulonephritis. Kidney Int 2003; 64: 509–518221. Reyes AA, Purkerson ML, Karl I, Klahr S. Dietary supple-

mentation with L-arginine ameliorates the progression of renal

disease in rats with subtotal nephrectomy. Am J Kidney Dis

1992; 20: 168–176

Treatment targets in renal fibrosis 3405

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 16: View - Nephrology Dialysis Transplantation

222. Peters H, Border WA, Ruckert M, Kramer S, Neumayer HH,Noble NA. L-arginine supplementation accelerates renalfibrosis and shortens life span in experimental lupus nephritis.Kidney Int 2003; 63: 1382–1392

223. Rangan GK, Wang Y, Harris DC. Pharmacologic modulatorsof nitric oxide exacerbate tubulointerstitial inflammation inproteinuric rats. J Am Soc Nephrol 2001; 12: 1696–1705

224. Chang B, Mathew R, Palmer LS, Valderrama E, Trachtman H.Nitric oxide in obstructive uropathy: role of endothelial nitricoxide synthase. J Urol 2002; 168: 1801–1804

225. Hochberg D, Johnson CW, Chen J et al. Interstitial fibrosis ofunilateral ureteral obstruction is exacerbated in kidneys of micelacking the gene for inducible nitric oxide synthase. Lab Invest2000; 80: 1721–1728

226. Ito K, Chen J, Khodadadian JJ et al. Liposome-mediatedtransfer of nitric oxide synthase gene improves renal function inureteral obstruction in rats. Kidney Int 2004; 66: 1365–75

227. Forbes MS, Thornhill BA, Park MH, Chevalier RL. Lack ofendothelial nitric-oxide synthase leads to progressive focal renalinjury. Am J Pathol 2007; 170: 87–99

228. Wang Y, Kramer S, Loof T et al. Stimulation of solubleguanylate cyclase slows progression in anti-thy1-inducedchronic glomerulosclerosis. Kidney Int 2005; 68: 47–61

229. Rodriguez-Iturbe B, Ferrebuz A, Vanegas V et al. Earlytreatment with cGMP phosphodiesterase inhibitor amelioratesprogression of renal damage. Kidney Int 2005; 68: 2131–2142

230. Lin SL, Chen RH, Chen YM et al. Pentoxifylline attenuatestubulointerstitial fibrosis by blocking Smad3/4-activated tran-scription and profibrogenic effects of connective tissue growthfactor. J Am Soc Nephrol 2005; 16: 2702–2713

231. Wang Y, Kramer S, Loof T et al. Enhancing cGMP inexperimental progressive renal fibrosis: soluble guanylatecyclase stimulation vs. phosphodiesterase inhibition. Am JPhysiol Renal Physiol 2006; 290: F167–F176

232. Lange-Sperandio B, Forbes MS, Thornhill B, Okusa MD,Linden J, Chevalier RL. A2A adenosine receptor agonist andPDE4 inhibition delays inflammation but fails to reduce injuryin experimental obstructive nephropathy. Nephron Exp Nephrol2005; 100: e113–e123

233. Tamada S, Asai T, Kuwabara N et al. Molecular mechanismsand therapeutic strategies of chronic renal injury: the role ofnuclear factor kappaB activation in the development of renalfibrosis. J Pharmacol Sci 2006; 100: 17–21

234. Kuwabara N, Tamada S, Iwai T et al. Attenuation of renalfibrosis by curcumin in rat obstructive nephropathy. Urology2006; 67: 440–446

235. Kanda T, Wakino S, Hayashi K, Homma K, Ozawa Y,Saruta T. Effect of fasudil on Rho-kinase and nephropathy insubtotally nephrectomized spontaneously hypertensive rats.Kidney Int 2003; 64: 2009–2019

236. Nagatoya K, Moriyama T, Kawada N et al. Y-27632 preventstubulointerstitial fibrosis in mouse kidneys with unilateralureteral obstruction. Kidney Int 2002; 61: 1684–1695

237. Nishikimi T, Akimoto K, Wang X et al. Fasudil, a Rho-kinaseinhibitor, attenuates glomerulosclerosis in Dahl salt-sensitiverats. J Hypertens 2004; 22: 1787–1796

238. Patel S, Takagi KI, Suzuki J et al. RhoGTPase activation is akey step in renal epithelial mesenchymal transdifferentiation.J Am Soc Nephrol 2005; 16: 1977–1984

239. Lai A, Frishman WH. Rho-kinase inhibition in the therapy ofcardiovascular disease. Cardiol Rev 2005; 13: 285–292

240. Fu P, Liu F, Su S et al. Signaling mechanism of renal fibrosis inunilateral ureteral obstructive kidney disease in ROCK1knockout mice. J Am Soc Nephrol 2006; 17: 3105–3114

241. Little MH. Regrow or repair: potential regenerative therapiesfor the kidney. J Am Soc Nephrol 2006; 17: 2390–2401

242. Prodromidi EI, Poulsom R, Jeffery R et al. Bone marrow-derived cells contribute to podocyte regeneration and ameliora-tion of renal disease in a mouse model of Alport syndrome.Stem Cells 2006; 24: 2448–2455

243. Ninichuk V, Gross O, Segerer S et al. Multipotent mesench-ymal stem cells reduce interstitial fibrosis but do not delayprogression of chronic kidney disease in collagen4A3-deficientmice. Kidney Int 2006; 70: 121–129

244. Sugimoto H, Mundel TM, Sund M, Xie L, Cosgrove D,Kalluri R. Bone-marrow-derived stem cells repair basementmembrane collagen defects and reverse genetic kidney disease.Proc Natl Acad Sci USA 2006; 103: 7321–7326

245. Broekema M, Harmsen MC, van Luyn MJ et al. Bone marrow-derived myofibroblasts contribute to the renal interstitialmyofibroblast population and produce procollagen I afterischemia/reperfusion in rats. J Am Soc Nephrol 2007; 18:165–175

246. Kunter U, Rong S, Boor P et al. Mesenchymal stem cellsprevent progressive experimental renal failure but maldiffer-entiate into glomerular adipocytes. J Am Soc Nephrol 2007; 18:1754–1764; doi:10.1681/ASN.2007010044

247. Akahori H, Ota T, Torita M, Ando H, Kaneko S, Takamura T.Tranilast prevents the progression of experimental diabeticnephropathy through suppression of enhanced extracellularmatrix gene expression. J Pharmacol Exp Ther 2005; 314:514–521

248. Kelly DJ, Zhang Y, Gow R, Gilbert RE. Tranilast attenuatesstructural and functional aspects of renal injury in the remnantkidney model. J Am Soc Nephrol 2004; 15: 2619–2629

249. Kelly DJ, Zhang Y, Cox AJ, Gilbert RE. Combination therapywith tranilast and angiotensin-converting enzyme inhibitionprovides additional renoprotection in the remnant kidneymodel. Kidney Int 2006; 69: 1954–1960

250. Soma J, Sato K, Saito H, Tsuchiya Y. Effect of tranilast inearly-stage diabetic nephropathy. Nephrol Dial Transplant2006; 21: 2795–2799

251. El-Koraie AF, Baddour NM, Adam AG, El Kashef EH, ElNahas AM. Role of stem cell factor and mast cells in theprogression of chronic glomerulonephritides. Kidney Int 2001;60: 167–172

252. Roberts IS, Brenchley PE. Mast cells: the forgotten cells ofrenal fibrosis. J Clin Pathol 2000; 53: 858–862

253. Kondo S, Kagami S, Kido H, Strutz F, Muller GA, Kuroda Y.Role of mast cell tryptase in renal interstitial fibrosis. J Am SocNephrol 2001; 12: 1668–1676

254. Kanamaru Y, Scandiuzzi L, Essig M et al. Mast cell-mediatedremodeling and fibrinolytic activity protect against fatalglomerulonephritis. J Immunol 2006; 176: 5607–5615

255. Miyazawa S, Hotta O, Doi N, Natori Y, Nishikawa K,Natori Y. Role of mast cells in the development of renal fibro-sis: use of mast cell-deficient rats. Kidney Int 2004; 65: 2228–37

256. Timoshanko JR, Kitching R, Semple TJ, Tipping PG,Holdsworth SR. A pathogenetic role for mast cells inexperimental crescentic glomerulonephritis. J Am Soc Nephrol2006; 17: 150–159

257. Lin L, Gerth AJ, Peng SL. Susceptibility of mast cell-deficientW/Wv mice to pristane-induced experimental lupus nephritis.Immunol Lett 2004; 91: 93–97

258. Silver RB, Reid AC, Mackins CJ et al. Mast cells: a uniquesource of renin.Proc Natl Acad Sci USA 2004; 101: 13607–13612

259. Lange-Sperandio B, Cachat F, Thornhill BA, Chevalier RL.Selectins mediate macrophage infiltration in obstructivenephropathy in newborn mice. Kidney Int 2002; 61: 516–524

260. Kuhlmann A, Haas CS, Gross ML et al. 1,25-Dihydroxyvitamin D3 decreases podocyte loss and podocytehypertrophy in the subtotally nephrectomized rat. Am J PhysiolRenal Physiol 2004; 286: F526–F533

261. Li Y, Spataro BC, Yang J, Dai C, Liu Y. 1,25-dihydroxyvitaminD inhibits renal interstitial myofibroblast activation byinducing hepatocyte growth factor expression. Kidney Int2005; 68: 1500–1510

262. Tan X, Li Y, Liu Y. Paricalcitol attenuates renal interstitialfibrosis in obstructive nephropathy. J Am Soc Nephrol 2006; 17:3382–3393

3406 P. Boor et al.

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022

Page 17: View - Nephrology Dialysis Transplantation

263. Zafiriou S, Stanners SR, Saad S, Polhill TS, Poronnik P,Pollock CA. Pioglitazone inhibits cell growth and reducesmatrix production in human kidney fibroblasts. J Am SocNephrol 2005; 16: 638–645

264. Li Y, Wen X, Spataro BC, Hu K, Dai C, Liu Y. hepatocytegrowth factor is a downstream effector that mediates theantifibrotic action of peroxisome proliferator-activated recep-tor-gamma agonists. J Am Soc Nephrol 2006; 17: 54–65

265. Panchapakesan U, Sumual S, Pollock CA, Chen X.PPARgamma agonists exert antifibrotic effects in renal tubularcells exposed to high glucose. Am J Physiol Renal Physiol 2005;289: F1153–F1158

266. Schaier M, Jocks T, Grone HJ, Ritz E, Wagner J. Retinoidagonist isotretinoin ameliorates obstructive renal injury. J Urol2003; 170: 1398–1402

267. Kang DH, Park EY, Yu ES, Lee YS, Yoon KI. Renoprotectiveeffect of erythropoietin (EPO): possibly via an amelioration ofrenal hypoxia with stimulation of angiogenesis in the kidney.Kidney Int 2005; 67: 1683

268. Lee SH, Li C, Lim SW et al. Attenuation of interstitialinflammation and fibrosis by recombinant human erythropoie-tin in chronic cyclosporine nephropathy. Am J Nephrol 2005;25: 64–76

269. Baggio B, Musacchio E, Priante G. Polyunsaturated fatty acidsand renal fibrosis: pathophysiologic link and potential clinicalimplications. J Nephrol 2005; 18: 362–367

270. Sebekova K, Eifert T, Klassen A, Heidland A, Amann K.Renal effects of S18886 (terutroban), a TP receptor antagonist,in an experimental model of type 2 diabetes. Diabetes 2007; 56:968–974

271. Shimizu MH, Coimbra TM, de Araujo M, Menezes LF,Seguro AC. N-acetylcysteine attenuates the progression ofchronic renal failure. Kidney Int 2005; 68: 2208–2217

272. Heller F, Lindenmeyer MT, Cohen CD et al. The contributionof B cells to renal interstitial inflammation. Am J Pathol 2007;170: 457–468

273. Kelly DJ, Chanty A, Gow RM, Zhang Y, Gilbert RE. Proteinkinase Cbeta inhibition attenuates osteopontin expression,macrophage recruitment, and tubulointerstitial injury inadvanced experimental diabetic nephropathy. J Am SocNephrol 2005; 16: 1654–1660

274. Kelly DJ, Zhang Y, Hepper C et al. Protein kinase C betainhibition attenuates the progression of experimental diabeticnephropathy in the presence of continued hypertension.Diabetes 2003; 52: 512–518

275. Meier M, Park JK, Overheu D et al. Deletion of protein kinaseC-{beta} isoform in vivo reduces renal hypertrophy but notalbuminuria in the streptozotocin-induced diabetic mousemodel. Diabetes 2007; 56: 346–354

276. Ohshiro Y, Ma RC, Yasuda Y et al. Reduction of diabetes-induced oxidative stress, fibrotic cytokine expression, and renaldysfunction in protein kinase Cbeta-null mice. Diabetes 2006;55: 3112–3120

277. Ma FY, Flanc RS, Tesch GH et al. A pathogenic role

for c-Jun amino-terminal kinase signaling in renalfibrosis and tubular cell apoptosis. J Am Soc Nephrol 2007;18: 472–484

278. Bohlender JM, Franke S, Stein G, Wolf G. Advanced glycationend products and the kidney. Am J Physiol Renal Physiol 2005;289: F645–F659

279. Li HY, Hou FF, Zhang X et al. Advanced oxidation proteinproducts accelerate renal fibrosis in a remnant kidney model.

J Am Soc Nephrol 2007; 18: 528–538280. Meldrum KK, Misseri R, Metcalfe P, Dinarello CA, Hile KL,

Meldrum DR. TNF-{alpha} neutralization amelioratesobstruction-induced renal fibrosis and dysfunction. Am JPhysiol Regul Integr Comp Physiol 2006; 292: R1456–R1464;doi: 10.1152/ajpregu.00620.2005

281. Chae YM, Park KK, Lee IK, Kim JK, Kim CH, Chang YC.Ring-Sp1 decoy oligonucleotide effectively suppresses extra-cellular matrix gene expression and fibrosis of rat kidney

induced by unilateral ureteral obstruction. Gene Ther 2006; 13:430–439

282. Park SH, Choi MJ, Song IK et al. Erythropoietin decreasesrenal fibrosis in mice with ureteral obstruction: role ofinhibiting tgf-beta-induced epithelial-to-mesenchymal transi-tion. J Am Soc Nephrol 2007; 18: 1497–1507

283. Matsumoto Y, Ueda S, Yamagishi S et al. Dimethylargininedimethylaminohydrolase prevents progression of renal

dysfunction by inhibiting loss of peritubular capillaries andtubulointerstitial fibrosis in a rat model of chronic kidneydisease. J Am Soc Nephrol 2007; 18: 1525–1533

284. Hudkins KL, Gilbertson DG, Carling M et al. ExogenousPDGF-D is a potent mesangial cell mitogen and causes a severe

mesangial proliferative glomerulopathy. J Am Soc Nephrol

2004; 15: 286–298285. Page-McCaw A, Ewald AJ, Werb Z. Matrix metalloproteinases

and the regulation of tissue remodelling. Nat Rev Mol Cell Biol

2007; 8: 221–233

Received for publication: 7.3.07Accepted in revised form: 25.5.07

Treatment targets in renal fibrosis 3407

Dow

nloaded from https://academ

ic.oup.com/ndt/article/22/12/3391/1913971 by guest on 04 February 2022