You are on page 1of 35

Articles in PresS. Am J Physiol Renal Physiol (January 6, 2016). doi:10.1152/ajprenal.00365.

2015

TGF- Signaling in the Kidney: Pro-fibrotic and Protective Effects

3
4

Angara Sureshbabu1*, Saif A. Muhsin2*, and Mary E. Choi1,2#

5
1

Joan and Sanford I. Weill Department of Medicine, Division of Nephrology & Hypertension,
Weill Cornell Medical College, and 2New York-Presbyterian Hospital-Weill Cornell Medical
Center, New York, NY 10065, USA

6
7
8
9
10
11
12

Running Title: Pro-fibrotic and Protective Effects of TGF-

13
14
15
16

*A. Sureshbabu and S.A. Muhsin contributed equally and are considered co-first authors

17
18

Address correspondence to:

19

Mary E. Choi, M.D.

20

Joan and Sanford I. Weill Department of Medicine

21

Division of Nephrology & Hypertension

22

Weill Cornell Medical College

23

525 East 68th Street, Box 3

24

New York, NY 10065

25

E-mail: mechoi@med.cornell.edu

1
Copyright 2016 by the American Physiological Society.

26

Abstract

27

Transforming Growth Factor-beta (TGF-) is generally considered as a central mediator of

28

fibrotic diseases. Indeed, much focus has been placed on inhibiting TGF- and its downstream

29

targets as ideal therapeutic strategies. However, pharmacological blockade of TGF- has not yet

30

translated into successful therapy for humans, which may be due to pleiotropic effects of TGF-

31

signaling. Equally, TGF- signaling as a protective response in kidney injury has been relatively

32

underexplored. Emerging body of evidence from experimental kidney disease models indicates

33

multifunctionality of TGF- capable of inducing pro-fibrotic and protective effects. This review

34

article discusses recent advances highlighting the diverse roles of TGF- in not only promoting

35

renal fibrosis, but also protective responses of TGF- signaling. We review, in particular,

36

growing evidence that supports protective effects of TGF- by mechanisms which include

37

inhibiting inflammation and induction of autophagy. Additional detailed studies are required to

38

fully understand the diverse mechanisms of TGF- actions in renal fibrosis and inflammation

39

that will likely direct towards effective anti-fibrotic therapies.

40
41

Keywords: TGF-, Smad, BMP, Kidney, Fibrosis, Autophagy, Apoptosis, Inflammation

42

43

Introduction

44

Transforming growth factor-beta (TGF-) is a multifunctional cytokine that is well

45

recognized to regulate a broad spectrum of cellular processes such as growth, differentiation,

46

apoptosis, wound repair, and the pathogenesis of fibrosis. It is a member of a superfamily of 38

47

cytokines that include TGF-, bone morphogenetic proteins (BMP), growth differentiation

48

factors, inhibins, and activins. TGF- superfamily members, in particular, TGF- and BMP have

49

been implicated to play important and diverse roles in chronic kidney disease (CKD) (71, 100).

50

The hallmark of progressive CKD is the excessive extracellular matrix (ECM) production

51

leading to renal fibrosis and TGF- has been generally considered a potent pro-fibrotic mediator

52

in this process (17, 48).

53

Mammalian TGF- includes three major isoforms that are encoded by distinct genes

54

(TGF-1, TGF-2, and TGF-3). The isoforms are synthesized as large precursor proteins (pro-

55

TGF-) forming dimeric complexes in the endoplasmic reticulum, and are subsequently cleaved

56

near the carboxy-terminus to yield mature 112 amino acid polypeptides, which share 60-80%

57

conservation across the three TGF- isoforms (99). The mature TGF- dimer remains associated

58

with cleaved latency peptide portion of the precursor as an inactive latent complex, and

59

dissociation from the complex results in biologically active TGF- (76). Thus, newly synthesized

60

TGF- bound to the latency associated peptide (LAP) forming a small latent complex (SLC) is

61

biologically inactive and cannot bind to its receptors. Through the formation of disulfide bonds,

62

this complex loosely binds to a latent TGF- binding protein (LTBP) to form a large latent

63

complex (LLC). TGF- is then secreted in a latent state and is stored in the ECM (2, 76).

64

Activation of TGF- involves release from the latent complex following exposure to a number of

65

different factors including integrins, proteases, metalloproteinases, reactive oxygen species

66

(ROS), plasmin, and acid, that allows binding to its cell surface receptors for initiation of TGF-

67

signaling (25, 76, 81, 87).

68

It is well established that activation of TGF- leads to pleiotropic cellular responses that

69

are mediated by TGF- signaling via interactions with TGF- receptor type I (TRI) and TGF-

70

receptor type II (TRII) and subsequent activation of intracellular Smad and non-Smad

71

dependent signaling pathways (13, 16, 57). In this review, we describe emerging body of

72

evidence from experimental kidney disease models demonstrating multifunctional nature of

73

TGF- capable of inducing pro-fibrotic and protective effects. We provide an overview on recent

74

advances highlighting the diverse roles of TGF- in not only promoting renal fibrosis but also

75

protective responses, focusing on growing evidence for protective effects of TGF- by

76

mechanisms which include inhibiting inflammation and induction of autophagy.

77
78

TGF- isoforms in the kidney

79

Studies of human kidney specimens have confirmed that the three major isoforms TGF-

80

1, TGF-2, and TGF-3 are expressed in the kidney (33). While functional redundancy between

81

the TGF- isoforms has been long recognized, there is a growing body of evidence for the

82

existence of non-redundant functions in inflammation and organ development (76). TGF-1, the

83

major focus of this review, is the predominant and best-characterized isoform, while TGF-2 and

84

TGF-3 are less known. Some clear differences among the isoforms have been noted. In the

85

normal human adult kidney, glomerular expression of TGF-2 and TGF-3 is seen mainly in

86

podocytes, whereas TGF-1 is primarily detected in the tubules but not in the glomeruli (33).

87

Interestingly, glomerular expression of TGF-1, generally with TGF-2 and TGF-3, was

88

detected in podocytes in kidney biopsy specimens from patients with proliferative

89

glomerulonephritis and in mesangial cells in diabetic nephropathy and IgA nephropathy (33).

90

Moreover, increased expression of TGF-1 was associated with development of severe

91

glomerulonephritis and glomerulosclerosis (33).

92

Biological actions of TGF- isoforms are mediated by ligand binding to its receptors for

93

the initiation of signaling. Both TGF-1 and TGF-3 bind directly with TRII, whereas TGF-2

94

requires the presence of a type III TGF- receptor (TRIII) for ligand binding to TRII (99).

95

Given the differences in the expression patterns and the mechanism of ligand binding, together

96

with apparent non-overlapping phenotypes of the three TGF- isoform knockout mice, it is not

97

unreasonable that some cellular responses may differ among the TGF- isoforms.

98

All three TGF- isoforms have been shown, in vitro, to induce ECM protein production

99

in various renal cells, including glomerular mesangial cells, renal fibroblasts, and renal tubular

100

epithelial cells (91, 99). While most studies have demonstrated similar pro-fibrotic effects of the

101

TGF- isoforms, a number of recent studies have suggested that TGF-2 and TGF-3 can exert

102

anti-fibrotic effects (72, 75, 96, 99). Studies in human podocytes demonstrated anti-fibrotic

103

effects of TGF-2 through upregulation of sphingosine kinase-1 (SK-1) activity, thereby

104

suppressing pro-fibrotic connective tissue growth factor (CTGF) expression (75). It is important

105

to note that similar anti-fibrotic effects of TGF-2 were not observed in other kidney cell types,

106

as all three isoforms equally induced upregulation of CTGF mRNA in cultured mesangial cells

107

and glomerular visceral epithelial cells (33). Moreover, TGF-2 stimulated the expression of

108

ECM proteins and induced EMT in tubular epithelial cells, whereas neutralizing antibody to

109

TGF-2 or repression of TGF-2 expression inhibited renal fibrogenesis (91). Further

110

investigations are warranted to clarify the seemingly opposite findings regarding the anti-fibrotic

111

roles of TGF-2 and TGF-3, which carry important implications for therapeutic targeting

112

strategy.

113
114

Activation of TGF-1 Signaling

115

TGF-1 signaling involves activation of complex intracellular networks to regulate

116

pleiotropic biological functions. As depicted in Figure 1, the active form of TGF-1 transmits

117

signals through type I and type II receptors, TRI and TRII. These cell surface receptors

118

contain serine/threonine kinases that phosphorylate transcription factor proteins called Smads to

119

initiate the canonical TGF- signaling pathway. TGF-1 ligand assembles a heteromeric

120

complex through binding to TRII, which then phosphorylates the kinase domain of TRI, and

121

leads to the downstream activation of the receptor-activated or regulatory Smads (R-Smads),

122

namely Smad2 and Smad3. The activated R-Smads form an oligomeric complex with the

123

common mediator Smad4 and undergo nuclear translocation to regulate transcription of target

124

genes (57). On the other hand, inhibitory Smads (Smad6 and Smad7) bind to activated TRI and

125

interfere with R-Smad activation. Smad7 has also been shown to negatively regulate TGF-

126

signaling through recruitment of E3 ubiquitin ligases, Smurf1 and Smurf2, which target TGF-

127

receptors and Smad7 to undergo degradation via proteasomal and lysosomal pathways (22, 36).

128

Additionally, TGF-1 signaling is mediated via non-canonical, non-Smad pathways

129

(Figure 1) including the mitogen-activated protein kinases (MAPKs), mediated by Ras-Raf

130

activating MEK (also called MAPKK) and extracellular signal-regulated kinase (ERK), (16, 68,

131

90) or TAK1 (TGF- activated kinase 1) pathway, the upstream MAPK kinase kinase (MKKK)

132

activating MKK3-p38 and MKK4-JNK (c-Jun N-terminal kinase) signaling cascades (13, 37, 38,

133

69). TGF-1 signaling also activates Rho family small GTPases, Rho, Rac, Cdc42 as well as

134

Phosphatidylinositol 3 Kinase (PI3K)/AKT and Integrin Linked Kinase (ILK) (3, 23, 50, 66, 95,

135

98).

136

Although the precise mechanisms underlying progressive renal fibrosis are not

137

completely understood, studies have shown that TGF-1, mediated via its canonical Smad

138

signaling pathway, drives development of renal fibrosis (77). Recent investigations have also

139

demonstrated that the non-canonical, non-Smad pathways can also mediate TGF-1 fibrogenic

140

responses (13, 38). However, new findings show that blocking TGF-1 signaling in renal cells in

141

vitro or in experimental animal models in vivo does not necessarily reduce renal fibrosis.

142
143

Anti-TGF- therapy in Renal Fibrosis

144

Based on significant pre-clinical evidence showing TGF-1 as a key mediator of fibrotic

145

diseases, much effort had been placed on inhibiting TGF-1 as a therapeutic strategy. Several

146

recent clinical trials using pharmacological blockade of TGF- in fibrotic kidney diseases are

147

summarized in Table 2. Therapeutic approach using neutralizing anti-TGF- antibody has been

148

explored in models of experimental diabetes induced by STZ and Leprdb/db diabetic mice,

149

demonstrating significant reduction in TGF-1 levels and inhibition of glomerular mesangial

150

matrix expansion with suppression of collagen and fibronectin expression (10, 80, 102).

151

However, the recent multicenter phase II trial sponsored by Eli Lilly using LY2382770, a

152

humanized neutralizing monoclonal antibody against TGF-1 for the treatment of diabetic

153

nephropathy had to be prematurely terminated due to futility in efficacy (clinicaltrials.gov:

154

NCT01113801). Fresolimumab (GC-1008, Genzyme) is a human monoclonal antibody that

155

neutralizes all three isoforms (TGF-1, 2, and 3). A single-dose infusion was shown to be

156

relatively safe and well tolerated in a phase I multicenter, open-label study conducted in patients

157

with treatment-resistant primary FSGS. A subsequent phase II multicenter, double-blind,

158

randomized study of fresolimumab in patients with steroid-resistant primary FSGS was recently

159

completed but no study results have been reported to date (clinicaltrials.gov: NCT01665391).

160

Pirfenidone is a small-molecular weight synthetic compound that has attracted much

161

attention as an orally active anti-fibrotic agent recently approved by the U.S. Food and Drug

162

Administration for the treatment of idiopathic pulmonary fibrosis (35). Pirfenidone effects are, in

163

part, mediated via inhibition of TGF-1 synthesis (78). In the kidney, the effects of pirfenidone

164

in suppressing renal fibrosis have been shown in several experimental models including diabetic

165

nephropathy, UUO, and subtotal nephrectomy (9, 74, 82). An open-label, single-center pilot

166

study demonstrated that pirfenidone significantly slowed renal function decline rate in patients

167

with FSGS, with a median improvement of 25% in patients who have moderate to severe CKD

168

and are already being treated with angiotensin antagonists (11). However, the phase II trial failed

169

to show an improvement in proteinuria (clinicaltrials.gov: NCT00001959). Limitations of the

170

trial included the small-scale, exploratory nature of the study and the lack of control group. A

171

phase I/II randomized, placebo-controlled trial conducted in 77 patients with overt diabetic

172

nephropathy who had albuminuria and reduced estimated glomerular filtration rate (eGFR) (20 to

173

75 ml/min per 1.73 m2) demonstrated encouraging results of pirfenidone in improving kidney

174

function, but without improvement in proteinuria (clinicaltrials.gov: NCT00063583). The mean

175

eGFR increased in the pirfenidone (1200 mg/d) group (+3.3 8.5 ml/min per 1.73 m), whereas,

176

the mean eGFR decreased in the placebo control group (-2.2 4.8 ml/min per 1.73 m) (79).

177

The results from these clinical trials portend some promise of pirfenidone as an anti-

178

fibrotic therapy to improve or slow the decline in kidney function associated with CKD. It should

179

be noted, however, that in the above studies, the assessment of kidney function was by eGFR

180

using the Modification of Diet in Renal Disease (MDRD) equation, and not by direct

181

measurement of GFR. In addition, no serial kidney biopsies were performed to confirm

182

histologic evidence of regression of renal fibrosis. Optimal dosage also may need to be further

183

identified as well.

184

Hence, anti-TGF- therapy has not yet translated into successful treatment in CKD

185

patients, and unfortunately, many of the clinical trials that have been completed to date have

186

been small and/or uncontrolled, and conclusive evidence regarding efficacy is limited and

187

disappointing. Below, we describe studies that provide evidence for pro-fibrotic effects as well

188

as evidence that supports protective effects of TGF-1.

189
190

Pro-fibrotic Effects of TGF-1

191

Renal fibrosis is characterized by excessive production and accumulation of ECM

192

proteins in the kidney that leads to parenchymal scarring, renal dysfunction, and ultimately end-

193

stage kidney disease. TGF-1 is the most extensively studied prototype member of the TGF-

194

superfamily in the context of fibrosis. TGF-1 is well known as a central mediator of renal

195

fibrosis and has long been considered to play potent roles in the progression of CKD (7, 17, 48).

196

Increased expression of TGF-1 mRNA and protein is seen in patients with fibrotic kidney

197

diseases, including IgA nephropathy, focal and segmental glomerulonephritis, lupus nephritis,

198

and diabetic and human immunodeficiency virusassociated nephropathy (5). Moreover, urinary

199

TGF-1 excretion is significantly elevated in patients with glomerular disease and heavy

200

proteinuria (24).

201

The overexpression of TGF-1 in transgenic mice with increased levels of circulating

202

active TGF-1 induces progressive renal disease characterized by mesangial expansion,

203

accumulation of ECM proteins, and development of glomerulosclerosis and tubulointerstitial

204

fibrosis (44). The accumulation of ECM proteins is stimulated by TGF-1 acting as both an

205

inducer of ECM synthesis and an inhibitor of ECM degradation by reduced synthesis of matrix

206

metalloproteinases, and increased synthesis of proteinase inhibitors, such as plasminogen

207

activator inhibitor-1 (PAI-1).

208

It has been shown that TGF-1 activation is required for its biological actions and

209

important in the development of renal fibrosis. Active TGF-1 is released when the latent

210

complex anchored to ECM is cleaved through proteolysis, following exposure to factors such as

211

integrins (2). Integrin v6 is a heterodimeric matrix receptor expressed in renal epithelia which

212

binds and activates latent TGF-1. Studies using an in vivo model of progressive renal fibrosis

213

induced by unilateral ureteral obstruction (UUO) in 6 integrin-null mice have demonstrated that

214

blockade of v6-mediated TGF-1 activation was associated with lower expression of type I

215

and type III collagen, PAI-1 and collagen content, and protected against tubulointerstitial fibrosis

216

(56). Thus, these studies provide evidence that local activation of TGF-1 is important in the

217

development of renal fibrosis.

218

A significant body of evidence exists in the literature that various approaches that disrupt

219

TGF-1 signaling protect against renal fibrosis. Inhibition of TGF-1 by using the proteoglycan

220

decorin which is a known inhibitor of TGF-1 or anti-TGF- antibodies abrogated development

221

of renal fibrosis (32, 62). BMP-7 is a TGF- superfamily member that has been shown to play a

222

protective role in renal fibrosis through anti-inflammatory, anti-oxidative, and anti-fibrotic

223

effects by counteracting the effects of TGF-1 (100). BMP-7 signaling pathway and its role in

224

fibrosis have been the subject of several recent reviews (6, 48, 71). In murine mesangial cells,

225

BMP-7 decreases Smad3 accumulation and inhibits the transcriptional upregulation of Smad3

10

226

targets such as plasminogen activator inhibitor-1 (PAI-1), thereby, suppresses the pro-fibrotic

227

effects of TGF-1 (93). Administration of exogenous recombinant human BMP-7 reduced renal

228

fibrosis in experimental models of renal injury in mice, including UUO and streptozotocin

229

(STZ)-induced diabetic kidney disease (64, 84, 92). Thus, BMP-7 opposes TGF-1/Smad3-

230

mediated renal fibrosis. Studies in Smad3-null mice revealed that targeted deletion of Smad3

231

signaling resulted in reduced ECM protein accumulation and attenuation of tubulointerstitial

232

fibrosis following UUO injury (31, 73, 77). They also showed that the numbers of

233

myofibroblasts, macrophages, and CD4/CD8 T cells, and monocyte influx into the kidney after

234

UUO were significantly decreased in the obstructed kidneys of Smad3-null mice (31, 77). These

235

findings suggest that TGF-1/Smad3 signaling mediates development of renal fibrosis and

236

inflammation.

237

TGF-1 signaling via the non-Smad pathways also participates in mediating pro-fibrotic

238

responses. Data from in vitro studies indicate that TGF-1-stimulated collagen expression in

239

mesangial cells is mediated via TAK1/MKK3/p38 signaling cascade, and fibronectin expression

240

in fibroblasts (13, 38, 69). Moreover, investigations using pharmacologic and genetic blockade in

241

in vivo models of glomerular and tubulointerstitial injury in mice have shown that the

242

TAK1/MKK3/p38 and JNK pathways promote renal fibrosis (13, 39). Both the p38 and JNK are

243

downstream targets of TAK1 activation. Conditional Tak1 gene deletion in mice suppressed

244

interstitial myofibroblast accumulation, collagen deposition, and expression of pro-fibrotic

245

molecules following UUO injury (55). These studies strongly suggest that the non-Smad

246

signaling pathway via TAK1 plays a critical role in ECM production and the pathogenesis of

247

kidney fibrosis.

248

11

249

Protective Effects of TGF-1

250

Given the abundance of evidence from the preclinical studies, targeting TGF- signaling

251

pathways seems logical in order to attenuate the development of renal fibrosis and progression of

252

CKD. However, contrary to highly anticipated results, data from clinical trials to date have not

253

been as strong as was hoped, and anti-TGF-1 treatment directed at pharmacological blockade of

254

TGF-1 has not yet translated into successful therapy for humans. Recently emerging evidence

255

suggests that it may be due, at least in part, to the fact that TGF-1 is capable of inducing not

256

only pro-fibrotic effects, but also protective effects (Table 1).

257

Numerous in vitro studies have shown that TGF-1 acts as a potent stimulator of ECM

258

production, including the synthesis of type I collagen and fibronectin. However, a recent report

259

by Neelisetty and colleagues demonstrated surprising findings that blocking TGF- signaling

260

through deletion of TRII in matrix-producing renal interstitial cells failed to suppress renal

261

fibrosis in mouse models kidney injury induced by UUO or aristolochic acid (67). The studies

262

showed that TRII deleted renal interstitial cells had significantly reduced type I collagen

263

production, but the overall renal fibrosis following kidney injury was not decreased in the

264

conditional knockout mice (67). These findings indicate that inhibiting TGF- signaling in

265

matrix-producing renal interstitial cells is not sufficient to protect against development of fibrosis

266

after kidney injury.

267

Interestingly, mice overexpressing latent (inactive) form of TGF-1 in keratinocytes

268

demonstrated protection against renal fibrosis in experimental obstructive kidney disease and

269

crescentic glomerulonephritis (28, 29). These mice had increased levels of latent, but not active,

270

TGF-1 in plasma and kidney tissue, and upregulation of renal Smad7. The observed protective

271

effects in the latent TGF-1 transgenic mice may be, at least in part, due to Smad7-mediated

12

272

inhibition of nuclear factor-B (NF-B) as well as inhibiting TGF-1 activation, consequently

273

protecting the kidney against inflammation and fibrosis.

274

Taken together, these studies suggest that TGF-1 is capable of exerting protective

275

effects by opposing the pro-fibrotic pathway via negative feedback effects of Smad2 and Smad7.

276

The maintenance of balance through this negative self-regulation of pro-fibrotic effects may be

277

important for tissue homeostasis. Under pathological conditions, it is possible that the balance

278

tips towards the pro-fibrotic pathway through dysregulation of the inhibitory mechanisms to

279

cause renal fibrosis, rather than solely through excess TGF-1, and that this may perhaps account

280

for the disappointing results of targeting TGF- in humans. Furthermore, there is emerging

281

evidence for the protective effects of TGF- by mechanisms that include inhibiting inflammation

282

and induction of autophagy, which will be discussed in the following sections.

283
284

TGF- Signaling and Renal Inflammation

285

Renal inflammation is implicated as a critical event in the initiation and progression of

286

renal fibrosis in CKD. Inflammatory response to renal injury involves infiltrating immune cells

287

as well as activated resident renal cells, which stimulate the production and release of pro-

288

fibrotic cytokines and growth factors and drive the pro-fibrotic process. Interestingly, despite the

289

established pro-inflammatory role, TGF-1 also possesses anti-inflammatory effects (49). Its

290

anti-inflammatory properties are evidenced by findings that targeted deletion of TGF-1 gene

291

results in profound multifocal inflammatory disease in mice (48, 83). Tgf-1-null mice develop a

292

rapid wasting syndrome and early death by 3-4 weeks of age, and display excessive

293

inflammatory responses with massive infiltration of lymphocytes and macrophages in many

294

organs (45). These data indicate that TGF-1 is a potent anti-inflammatory molecule.

13

295

It has been proposed that the reason why therapies directed against TGF-1 in general

296

have yielded disappointing results is because blockade of TGF-1 signaling not only abrogates

297

its pro-fibrotic effects, but also inhibits its anti-inflammatory effects, and thereby can promote

298

renal fibrosis. Indeed, in vivo studies in mice have confirmed that conditional deletion of TRII

299

from the kidney tubular epithelial cells enhanced NF-B signaling and renal inflammation, with

300

upregulation of pro-inflammatory cytokines such as interleukin-1 (IL-1) and tumor necrosis

301

factor- (TNF-) in the kidney following UUO injury (59). Similarly, disruption of TRII in

302

cultured renal tubular epithelial cells and fibroblasts resulted in impairment of the anti-

303

inflammatory effect of TGF-1 (59). Furthermore, conditional Smad4 knockout mice in which

304

Smad4 was specifically deleted from the kidney tubular epithelial cells displayed significantly

305

enhanced renal inflammation as evidenced by increased infiltration of CD45+ leukocytes and

306

F4/80+ macrophages, and upregulation of pro-inflammatory cytokines IL-1, TNF-, monocyte

307

chemoattractant protein-1 (MCP-1), as well as intercellular adhesion molecule-1 (ICAM-1) in

308

the UUO kidney (60). Thus, inhibition of TGF- signaling through either disruption of TRII or

309

Smad4 resulted in enhanced renal inflammation, suggesting that TGF- possesses anti-

310

inflammatory effects in the kidney.

311

Further evidence indicates that loss of Smad4 represses Smad7 transcription that leads to

312

decreased IB (nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor,

313

alpha) expression but enhanced NF-B activation in the tubulointerstitium after UUO, thereby

314

promoting renal inflammation (60). Moreover, Smad7 knockout mice display enhanced renal

315

inflammation with increased NF-B/p65 phosphorylation and increased macrophage infiltration

316

in models of UUO and STZ-induced diabetes (8, 14). In contrast, Smad7 gene transfer into the

317

kidney of STZ-induced diabetic rats using an ultrasound microbubble-mediated technique

14

318

significantly attenuated NF-kB/p65-driven renal inflammation and macrophage infiltration in

319

diabetic rats (8). Latent TGF-1 overexpression in mice also protected against renal

320

inflammation, via up-regulation of renal Smad7 and IB and suppression of NF-B activation

321

in models of UUO and crescentic glomerulonephritis (29, 94). Thus, Smad7-mediated inhibition

322

of NF-B activation via the induction of IB may be a central mechanism to abrogate renal

323

inflammation (Figure 2).

324

Recent investigations have also explored the role of TGF- signaling and macrophage

325

differentiation in inflammation. Kidney injury stimulates the recruitment and induction of

326

macrophages to undergo classical activation and differentiate into pro-inflammatory M1 and

327

anti-inflammatory M2 macrophages (1, 47). More recently, M2 macrophages have been further

328

subdivided into three subsets namely M2a, M2b and M2c macrophages. Regulatory M2c

329

macrophages are induced by TGF- or IL-10 and recent evidence reported that this particular

330

subset displayed protection against renal fibrosis (54, 65). While M2c phenotype is induced by

331

TGF- and generally considered to be anti-inflammatory, M2c macrophages in turn produce

332

TGF-, and it has been proposed that this might be a mechanism by which macrophages drive

333

the development and progression of fibrosis (54). However, a recent study using conditional

334

deletion of TGF-1 in macrophages suggest that macrophage-derived TGF- may not serve as a

335

functionally important source of TGF- for renal fibrosis (30).

336
337

TGF- Signaling and Autophagy

338

Recent evidence proposes a new mechanism by which TGF-1 can provide

339

cytoprotective effects via induction of a highly conserved cellular process known as

340

macroautophagy, hereafter referred to as autophagy. It has long been known that autophagy is an

15

341

intracellular process that occurs in all eukaryotes and results in the degradation of sequestered

342

cytoplasmic elements in lysosomes (12). The execution of autophagy begins with the

343

establishment of protein complexes that function in the formation of the phagophore, also known

344

as the isolation membrane. The next major step, elongation of the autophagosome includes two

345

ubiquitin-like protein complexes, namely autophagy-related gene (ATG)5 ATG12 and

346

microtubule-associated protein 1 light chain 3 (LC3) conjugation systems. These two major steps

347

are sequentially followed by autophagosome-lysosomal fusion, lysosomal degradation and

348

release of free amino acids and fatty acids (12, 85, 86).

349

Accumulating data implicate critical roles of autophagy in health and kidney disease (17).

350

TGF-1 stimulation induced the accumulation of autophagosomes and conversion of LC3 to the

351

lipidated form, LC3-II and upregulated autophagy-related genes, ATG5, ATG7, LC3 and Beclin

352

1 in renal tubular epithelial cells (19, 42, 97). We also recently reported that TGF-1 induced

353

autophagy in glomerular mesangial cells (18, 40). Hence, these studies provide strong evidence

354

that TGF-1 acts as an inducer of autophagy.

355

Important advances have also been made in our understanding of the mechanisms of

356

autophagy activation by TGF-1 signaling pathways (18, 40, 88). In primary mesangial cells, we

357

further showed evidence that TGF-1 induces autophagy through the TAK1-MKK3-p38

358

signaling pathway, and autophagy promotes intracellular degradation of collagen and aggregated,

359

insoluble procollagen (40). Moreover, TGF-1 protects against serum deprivation-induced

360

mesangial cell apoptosis through the induction of autophagy via TAK1 and AKT activation (18).

361

The role of AKT-mTOR signaling pathway has also been demonstrated to mediate the activation

362

of autophagy in vivo following UUO injury in rats (41).

16

363

Autophagy is increasingly recognized as an important cellular defense against various

364

stress stimuli, and dysregulated autophagy is implicated in diseases characterized by progressive

365

kidney fibrosis (17). Upregulation of autophagy expression was noted in human kidney biopsies

366

of patients with acquired proteinuric diseases such as focal segmental glomerulosclerosis (FSGS)

367

(27). We and others have demonstrated induction of autophagy following UUO-induced kidney

368

injury (19, 40, 41). Using GFP-LC3 transgenic mice, we confirmed that autophagy is induced in

369

renal tubular epithelial cells of obstructed kidneys after UUO (19). Autophagy deficiency led to

370

enhanced kidney fibrosis following UUO injury. For instance, mice deficient in autophagic

371

protein Beclin 1 by heterozygous deletion of Beclin 1 displayed increased collagen deposition in

372

the kidney (40). Both Lc3b-null mice and Beclin 1-heterozygous mice displayed enhanced

373

collagen deposition in the obstructed kidneys after UUO (19). Treatment with autophagy

374

inhibitor 3-Methyladenine (3-MA) also enhanced renal tubular cell apoptosis and

375

tubulointerstitial fibrosis in the obstructed kidneys after UUO in rats (41).

376

Podocytes exhibit a high basal level of autophagy, important in cell survival, given that

377

they are terminally differentiated and have a very limited capacity for cell division and

378

replacement (27). Podocyte-specific deletion of the Atg5 gene resulted in increased susceptibility

379

to injury with more severe albuminuria, foot process effacement, loss of podocytes, and

380

glomerulosclerosis, following exposure to inducers of proteinuric glomerular injury, such as

381

puromycin aminonucleoside, adriamycin, or LPS (27). Induction of autophagy has also been

382

shown to be protective against cyclosporine-induced renal tubular cell death (70). Collectively,

383

these findings provide evidence that TGF-1-induced autophagy provides cytoprotective effects

384

that promote renal cell survival and negatively regulate and limit ECM accumulation in the

385

kidney, and thereby protect against kidney fibrosis (Figure 3).

17

386
387

Negative Feedback Regulation of TGF- Signaling

388

The diverse roles of TGF-1 necessitate that its signaling is tightly regulated at various

389

levels. TGF-1 induces Smad7, which is an inhibitory Smad that acts in a negative feedback loop

390

to negatively regulate TGF-1 (22). Overexpression of Smad7 has been shown to oppose TGF-

391

1-mediated renal fibrosis following UUO or STZ-induced diabetes, whereas deficiency of

392

Smad7 aggravated the severity of the renal fibrosis (8, 14, 46). Smad7-null mice display

393

enhanced TGF-/Smad3 signaling and worsened progressive renal fibrosis in a model of

394

angiotensin II-mediated hypertensive nephropathy (51), whereas treatment with Smad7

395

prevented angiotensin II-induced hypertensive nephropathy (52). Disruption of Smad7 also

396

exacerbated chronic aristolochic acid nephropathy (AAN) in mice, a progressive CKD related to

397

herb medicine (15). Furthermore, overexpression of Smad7 in the kidneys with established

398

chronic AAN attenuated progression of chronic AAN through inactivation of TGF-/Smad3

399

signaling (15).

400

Similarly, studies utilizing other approaches to inhibit TGF- signaling, downstream of

401

the cell-surface expressed TGF- receptors, have also shown failure to protect against kidney

402

fibrosis. Findings in mice with targeted conditional deletion of Smad2 in the kidney show that

403

blockade of Smad2 signaling pathway not only failed to protect against the development of

404

fibrosis after UUO injury but actually enhanced renal fibrosis (58). Moreover, knockdown of

405

Smad2 in cultured renal tubular epithelial cells and fibroblasts resulted in enhanced ECM

406

production in response to TGF-1 stimulation, with increased expression of type I and type III

407

collagen, and TIMP-1, and decreased expression of MMP-2, a major matrix-degrading enzyme

408

(58). Smad2 deletion was associated with increases in Smad3 phosphorylation, nuclear

18

409

translocation, and Smad3 binding to collagen (COL1A2) promoter, and autoinduction of TGF-1

410

(58). These studies indicate that inhibition of Smad2 promotes renal fibrosis via enhanced Smad3

411

signaling. On the other hand, overexpression of Smad2 resulted in attenuation of TGF-1-

412

induced Smad3 phosphorylation and type I collagen production in renal tubular epithelial cells

413

(58). Taken together, these findings suggest that Smad2 functions to protect against development

414

of renal fibrosis through countering the pro-fibrotic function of Smad3 signaling.

415

Our recent investigations have uncovered a novel mechanism that autophagy regulates

416

TGF-1 expression and suppresses kidney fibrosis induced by UUO. Increased levels of mature

417

TGF-1 were seen in the obstructed kidneys of LC3 deficient mice upon UUO injury (19).

418

Similarly, mature TGF-1 levels were increased in LC3 deficient renal tubular epithelial cells,

419

and in cells treated with autophagy inhibitor bafilomycin A1, without alterations in TGF-1

420

mRNA (19). These findings suggest a novel feedback mechanism to limit TGF- signaling

421

through autophagic degradation and suppress kidney fibrosis.

422

A number of endogenous modulators of TGF- signaling has been described, such as

423

secreted Klotho, which may serve as a promising therapeutic target against renal fibrosis. Klotho

424

is a transmembrane protein expressed in renal tubular epithelial cells, and its extracellular

425

domain is secreted by ectodomain shedding (4). Secreted Klotho inhibited TGF-1-induced EMT

426

in cultured cells, and administration of secreted Klotho protein to mice suppressed renal fibrosis

427

induced by UUO (20). Interestingly, it has been suggested that the Klotho-mediated

428

renoprotective effects may be, at least in part, due to upregulated autophagy flux (4). Secreted

429

Klotho protein directly binds to TRII, thereby inhibiting TGF-1 binding to cell surface

430

receptors, and inhibit TGF-1 signaling (20). In contrast, high plasma levels of Lipoprotein(a)

431

[Lp(a)], a low-density lipoprotein (LDL)-like particle, can inhibit the activation of latent TGF-1

19

432

by competing with the binding of plasminogen to cell or matrix surfaces (26, 43). The

433

proteoglycan decorin is another natural antagonist of TGF-1 that binds active TGF-1 and can

434

neutralize the biological effects of TGF-1 (32). Retinoic acid, particularly all-trans retinoic acid

435

(ATRA), has also been shown to suppress renal expression of TGF-1 and TRII, leading to

436

decreased ECM accumulation and inhibit progression of renal fibrosis (53, 63, 101). Other

437

studies have indicated that ATRA exacerbates kidney injury with increased glomerular ECM,

438

mesangial cell activation, glomerular macrophage influx, and immune complex deposition in a

439

model of membranoproliferative glomerulonephritis in mice (34, 101). Thus, the therapeutic

440

effects of retinoic acid in fibrotic kidney disease remain somewhat controversial, suggesting that

441

caution is needed in applying retinoid therapy to human disease. No published clinical studies

442

using soluble Kotho protein or ATRA administration in patients with CKD have been reported to

443

date, despite the preclinical data supporting their therapeutic potential. Recently, a phase I/II trial

444

has been initiated to evaluate the safety and efficacy of isotretinoin (13-cis retinoic acid)

445

treatment in patients with biopsy-proven FSGS, minimal change disease, or collapsing

446

glomerulopathy (clinicaltrials.gov: NCT00098020). The results of this clinical study will be of

447

great interest.

448
449

Concluding Remarks

450

Renal fibrosis represents the common pathway in kidney injury of most progressive CKD

451

leading to renal failure and end-stage kidney disease. Unfortunately, there are no effective

452

therapies that prevent progressive renal fibrosis. Current treatment for patients with CKD

453

primarily relies on inhibition of the renin-angiotensin-aldosterone system (RAAS) by

454

angiotensin-converting enzyme (ACE) inhibitors and angiotensin II receptor blockers (ARBs)

20

455

(61). However, the magnitude of the renoprotective effects of RAAS blockade is estimated to be

456

only about a 20% risk reduction of progression (21).

457

TGF-1 remains an attractive target for treating fibrotic kidney diseases, to counter its

458

potent pro-fibrotic effects. However, as this review highlights its opposing protective effects,

459

indiscriminate complete blockade of TGF- functions is not sufficient to reduce fibrosis, and

460

may actually aggravate the disease in some pathological settings. Perhaps, there are also

461

interspecies differences suggesting that results derived from preclinical studies in mice and in

462

vitro may not necessarily translate to humans. TGF-1 actions depend on multiple factors

463

including the involved cell type and context, the dose, isoform, and the distinct Smad and non-

464

Smad signaling pathways. Therefore, therapeutic targeting of TGF-1 requires more optimal

465

strategies such as those selectively directed at specific signaling pathway(s), and future

466

investigations to better understand the precise molecular mechanisms of TGF-1 signaling are

467

warranted.

468
469
470
471

Acknowledgements

472

This work was supported in part by the National Institutes of Health grants R01 DK57661 and

473

R01 HL079904 to M.E.C.

474

21

475

References

476
477
478
479
480
481
482
483
484
485
486
487
488
489
490
491
492
493
494
495
496
497
498
499
500
501
502
503
504
505
506
507
508
509
510
511
512
513
514
515
516
517
518
519

1.
Anders HJ, Ryu M. Renal microenvironments and macrophage phenotypes determine
progression or resolution of renal inflammation and fibrosis. Kidney Int 80: 915-925, 2011.
2.
Annes JP, Munger JS, Rifkin DB. Making sense of latent TGFbeta activation. J Cell
Sci 116: 217-224, 2003.
3.
Bakin AV, Tomlinson AK, Bhowmick NA, Moses HL, Arteaga CL.
Phosphatidylinositol 3-kinase function is required for transforming growth factor beta-mediated
epithelial to mesenchymal transition and cell migration. J Biol Chem 275: 36803-36810, 2000.
4.
Bian A, Neyra JA, Zhan M, Hu MC. Klotho, stem cells, and aging. Clin Interv Aging
10: 1233-1243, 2015.
5.
Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor beta in
human disease. N Engl J Med 342: 1350-1358, 2000.
6.
Boon MR, van der Horst G, van der Pluijm G, Tamsma JT, Smit JW, Rensen PC.
Bone morphogenetic protein 7: a broad-spectrum growth factor with multiple target therapeutic
potency. Cytokine Growth Factor Rev 22: 221-229, 2011.
7.
Border WA, Noble NA. Transforming growth factor beta in tissue fibrosis. N Engl J
Med 331: 1286-1292, 1994.
8.
Chen HY, Huang XR, Wang W, Li JH, Heuchel RL, Chung AC, Lan HY. The
protective role of Smad7 in diabetic kidney disease: mechanism and therapeutic potential.
Diabetes 60: 590-601, 2011.
9.
Chen JF, Ni HF, Pan MM, Liu H, Xu M, Zhang MH, Liu BC. Pirfenidone inhibits
macrophage infiltration in 5/6 nephrectomized rats. Am J Physiol Renal Physiol 304: F676-685,
2013.
10.
Chen S, Iglesias-de la Cruz MC, Jim B, Hong SW, Isono M, Ziyadeh FN.
Reversibility of established diabetic glomerulopathy by anti-TGF-beta antibodies in db/db mice.
Biochem Biophys Res Commun 300: 16-22, 2003.
11.
Cho ME, Smith DC, Branton MH, Penzak SR, Kopp JB. Pirfenidone slows renal
function decline in patients with focal segmental glomerulosclerosis. Clin J Am Soc Nephrol 2:
906-913, 2007.
12.
Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. N Engl J Med
368: 651-662, 2013.
13.
Choi ME, Ding Y, Kim SI. TGF-beta signaling via TAK1 pathway: role in kidney
fibrosis. Semin Nephrol 32: 244-252, 2012.
14.
Chung AC, Huang XR, Zhou L, Heuchel R, Lai KN, Lan HY. Disruption of the
Smad7 gene promotes renal fibrosis and inflammation in unilateral ureteral obstruction (UUO) in
mice. Nephrol Dial Transplant 24: 1443-1454, 2009.
15.
Dai XY, Zhou L, Huang XR, Fu P, Lan HY. Smad7 protects against chronic
aristolochic acid nephropathy in mice. Oncotarget 2015.
16.
Derynck R, Zhang YE. Smad-dependent and Smad-independent pathways in TGF-beta
family signalling. Nature 425: 577-584, 2003.
17.
Ding Y, Choi ME. Regulation of autophagy by TGF-beta: emerging role in kidney
fibrosis. Semin Nephrol 34: 62-71, 2014.
18.
Ding Y, Kim JK, Kim SI, Na HJ, Jun SY, Lee SJ, Choi ME. TGF-{beta}1 protects
against mesangial cell apoptosis via induction of autophagy. J Biol Chem 285: 37909-37919,
2010.

22

520
521
522
523
524
525
526
527
528
529
530
531
532
533
534
535
536
537
538
539
540
541
542
543
544
545
546
547
548
549
550
551
552
553
554
555
556
557
558
559
560
561
562
563
564
565

19.
Ding Y, Kim SL, Lee SY, Koo JK, Wang Z, Choi ME. Autophagy regulates TGF-beta
expression and suppresses kidney fibrosis induced by unilateral ureteral obstruction. J Am Soc
Nephrol 25: 2835-2846, 2014.
20.
Doi S, Zou Y, Togao O, Pastor JV, John GB, Wang L, Shiizaki K, Gotschall R,
Schiavi S, Yorioka N, Takahashi M, Boothman DA, Kuro-o M. Klotho inhibits transforming
growth factor-beta1 (TGF-beta1) signaling and suppresses renal fibrosis and cancer metastasis in
mice. J Biol Chem 286: 8655-8665, 2011.
21.
Drawz PE, Rosenberg ME. Slowing progression of chronic kidney disease. Kidney Int
Suppl (2011) 3: 372-376, 2013.
22.
Ebisawa T, Fukuchi M, Murakami G, Chiba T, Tanaka K, Imamura T, Miyazono
K. Smurf1 interacts with transforming growth factor-beta type I receptor through Smad7 and
induces receptor degradation. J Biol Chem 276: 12477-12480, 2001.
23.
Edlund S, Landstrom M, Heldin CH, Aspenstrom P. Transforming growth factorbeta-induced mobilization of actin cytoskeleton requires signaling by small GTPases Cdc42 and
RhoA. Mol Biol Cell 13: 902-914, 2002.
24.
Goumenos DS, Tsakas S, El Nahas AM, Alexandri S, Oldroyd S, Kalliakmani P,
Vlachojannis JG. Transforming growth factor-beta(1) in the kidney and urine of patients with
glomerular disease and proteinuria. Nephrol Dial Transplant 17: 2145-2152, 2002.
25.
Grabias BM, Konstantopoulos K. The physical basis of renal fibrosis: effects of altered
hydrodynamic forces on kidney homeostasis. Am J Physiol Renal Physiol 306: F473-485, 2014.
26.
Grainger DJ, Kirschenlohr HL, Metcalfe JC, Weissberg PL, Wade DP, Lawn RM.
Proliferation of human smooth muscle cells promoted by lipoprotein(a). Science 260: 1655-1658,
1993.
27.
Hartleben B, Godel M, Meyer-Schwesinger C, Liu S, Ulrich T, Kobler S, Wiech T,
Grahammer F, Arnold SJ, Lindenmeyer MT, Cohen CD, Pavenstadt H, Kerjaschki D,
Mizushima N, Shaw AS, Walz G, Huber TB. Autophagy influences glomerular disease
susceptibility and maintains podocyte homeostasis in aging mice. J Clin Invest 120: 1084-1096,
2010.
28.
Huang XR, Chung AC, Wang XJ, Lai KN, Lan HY. Mice overexpressing latent TGFbeta1 are protected against renal fibrosis in obstructive kidney disease. Am J Physiol Renal
Physiol 295: F118-127, 2008.
29.
Huang XR, Chung AC, Zhou L, Wang XJ, Lan HY. Latent TGF-beta1 protects
against crescentic glomerulonephritis. J Am Soc Nephrol 19: 233-242, 2008.
30.
Huen SC, Moeckel GW, Cantley LG. Macrophage-specific deletion of transforming
growth factor-beta1 does not prevent renal fibrosis after severe ischemia-reperfusion or
obstructive injury. Am J Physiol Renal Physiol 305: F477-484, 2013.
31.
Inazaki K, Kanamaru Y, Kojima Y, Sueyoshi N, Okumura K, Kaneko K,
Yamashiro Y, Ogawa H, Nakao A. Smad3 deficiency attenuates renal fibrosis,
inflammation,and apoptosis after unilateral ureteral obstruction. Kidney Int 66: 597-604, 2004.
32.
Isaka Y, Brees DK, Ikegaya K, Kaneda Y, Imai E, Noble NA, Border WA. Gene
therapy by skeletal muscle expression of decorin prevents fibrotic disease in rat kidney. Nat Med
2: 418-423, 1996.
33.
Ito Y, Goldschmeding R, Kasuga H, Claessen N, Nakayama M, Yuzawa Y, Sawai A,
Matsuo S, Weening JJ, Aten J. Expression patterns of connective tissue growth factor and of
TGF-beta isoforms during glomerular injury recapitulate glomerulogenesis. Am J Physiol Renal
Physiol 299: F545-558, 2010.

23

566
567
568
569
570
571
572
573
574
575
576
577
578
579
580
581
582
583
584
585
586
587
588
589
590
591
592
593
594
595
596
597
598
599
600
601
602
603
604
605
606
607
608
609
610

34.
Iyoda M, Hudkins KL, Wietecha TA, Banas MC, Guo S, Liu G, Wang L,
Kowalewska J, Alpers CE. All-trans-retinoic acid aggravates cryoglobulin-associated
membranoproliferative glomerulonephritis in mice. Nephrol Dial Transplant 22: 3451-3461,
2007.
35.
Karimi-Shah BA, Chowdhury BA. Forced vital capacity in idiopathic pulmonary
fibrosis--FDA review of pirfenidone and nintedanib. N Engl J Med 372: 1189-1191, 2015.
36.
Kavsak P, Rasmussen RK, Causing CG, Bonni S, Zhu H, Thomsen GH, Wrana JL.
Smad7 binds to Smurf2 to form an E3 ubiquitin ligase that targets the TGF beta receptor for
degradation. Mol Cell 6: 1365-1375, 2000.
37.
Kim SI, Kwak JH, Na HJ, Kim JK, Ding Y, Choi ME. Transforming growth factorbeta (TGF-beta1) activates TAK1 via TAB1-mediated autophosphorylation, independent of
TGF-beta receptor kinase activity in mesangial cells. J Biol Chem 284: 22285-22296, 2009.
38.
Kim SI, Kwak JH, Zachariah M, He Y, Wang L, Choi ME. TGF-beta-activated
kinase 1 and TAK1-binding protein 1 cooperate to mediate TGF-beta1-induced MKK3-p38
MAPK activation and stimulation of type I collagen. Am J Physiol Renal Physiol 292: F14711478, 2007.
39.
Kim SI, Lee SY, Wang Z, Ding Y, Haque N, Zhang J, Zhou J, Choi ME. TGF-betaactivated kinase 1 is crucial in podocyte differentiation and glomerular capillary formation. J Am
Soc Nephrol 25: 1966-1978, 2014.
40.
Kim SI, Na HJ, Ding Y, Wang Z, Lee SJ, Choi ME. Autophagy promotes intracellular
degradation of type I collagen induced by transforming growth factor (TGF)-beta1. J Biol Chem
287: 11677-11688, 2012.
41.
Kim WY, Nam SA, Song HC, Ko JS, Park SH, Kim HL, Choi EJ, Kim YS, Kim J,
Kim YK. The role of autophagy in unilateral ureteral obstruction rat model. Nephrology
(Carlton) 17: 148-159, 2012.
42.
Koesters R, Kaissling B, Lehir M, Picard N, Theilig F, Gebhardt R, Glick AB,
Hahnel B, Hosser H, Grone HJ, Kriz W. Tubular overexpression of transforming growth
factor-beta1 induces autophagy and fibrosis but not mesenchymal transition of renal epithelial
cells. Am J Pathol 177: 632-643, 2010.
43.
Kojima S, Harpel PC, Rifkin DB. Lipoprotein (a) inhibits the generation of
transforming growth factor beta: an endogenous inhibitor of smooth muscle cell migration. J Cell
Biol 113: 1439-1445, 1991.
44.
Kopp JB, Factor VM, Mozes M, Nagy P, Sanderson N, Bottinger EP, Klotman PE,
Thorgeirsson SS. Transgenic mice with increased plasma levels of TGF-beta 1 develop
progressive renal disease. Lab Invest 74: 991-1003, 1996.
45.
Kulkarni AB, Huh CG, Becker D, Geiser A, Lyght M, Flanders KC, Roberts AB,
Sporn MB, Ward JM, Karlsson S. Transforming growth factor beta 1 null mutation in mice
causes excessive inflammatory response and early death. Proc Natl Acad Sci U S A 90: 770-774,
1993.
46.
Lan HY, Mu W, Tomita N, Huang XR, Li JH, Zhu HJ, Morishita R, Johnson RJ.
Inhibition of renal fibrosis by gene transfer of inducible Smad7 using ultrasound-microbubble
system in rat UUO model. J Am Soc Nephrol 14: 1535-1548, 2003.
47.
Lee S, Huen S, Nishio H, Nishio S, Lee HK, Choi BS, Ruhrberg C, Cantley LG.
Distinct macrophage phenotypes contribute to kidney injury and repair. J Am Soc Nephrol 22:
317-326, 2011.

24

611
612
613
614
615
616
617
618
619
620
621
622
623
624
625
626
627
628
629
630
631
632
633
634
635
636
637
638
639
640
641
642
643
644
645
646
647
648
649
650
651
652
653
654
655
656

48.
Lee SY, Kim SI, Choi ME. Therapeutic targets for treating fibrotic kidney diseases.
Transl Res 165: 512-530, 2015.
49.
Li MO, Wan YY, Sanjabi S, Robertson AK, Flavell RA. Transforming growth factorbeta regulation of immune responses. Annu Rev Immunol 24: 99-146, 2006.
50.
Li Y, Tan X, Dai C, Stolz DB, Wang D, Liu Y. Inhibition of integrin-linked kinase
attenuates renal interstitial fibrosis. J Am Soc Nephrol 20: 1907-1918, 2009.
51.
Liu GX, Li YQ, Huang XR, Wei L, Chen HY, Shi YJ, Heuchel RL, Lan HY.
Disruption of Smad7 promotes ANG II-mediated renal inflammation and fibrosis via Sp1-TGFbeta/Smad3-NF.kappaB-dependent mechanisms in mice. PLoS One 8: e53573, 2013.
52.
Liu GX, Li YQ, Huang XR, Wei LH, Zhang Y, Feng M, Meng XM, Chen HY, Shi
YJ, Lan HY. Smad7 inhibits AngII-mediated hypertensive nephropathy in a mouse model of
hypertension. Clin Sci (Lond) 127: 195-208, 2014.
53.
Long YB, Qin YH, Zhou TB, Lei FY. Association of retinoic acid receptors with
extracellular matrix accumulation in rats with renal interstitial fibrosis disease. Int J Mol Sci 13:
14073-14085, 2012.
54.
Lu J, Cao Q, Zheng D, Sun Y, Wang C, Yu X, Wang Y, Lee VW, Zheng G, Tan TK,
Wang X, Alexander SI, Harris DC, Wang Y. Discrete functions of M2a and M2c macrophage
subsets determine their relative efficacy in treating chronic kidney disease. Kidney Int 84: 745755, 2013.
55.
Ma FY, Tesch GH, Ozols E, Xie M, Schneider MD, Nikolic-Paterson DJ. TGF-beta1activated kinase-1 regulates inflammation and fibrosis in the obstructed kidney. Am J Physiol
Renal Physiol 300: F1410-1421, 2011.
56.
Ma LJ, Yang H, Gaspert A, Carlesso G, Barty MM, Davidson JM, Sheppard D,
Fogo AB. Transforming growth factor-beta-dependent and -independent pathways of induction
of tubulointerstitial fibrosis in beta6(-/-) mice. Am J Pathol 163: 1261-1273, 2003.
57.
Massague J. TGFbeta signalling in context. Nat Rev Mol Cell Biol 13: 616-630, 2012.
58.
Meng XM, Huang XR, Chung AC, Qin W, Shao X, Igarashi P, Ju W, Bottinger EP,
Lan HY. Smad2 protects against TGF-beta/Smad3-mediated renal fibrosis. J Am Soc Nephrol
21: 1477-1487, 2010.
59.
Meng XM, Huang XR, Xiao J, Chen HY, Zhong X, Chung AC, Lan HY. Diverse
roles of TGF-beta receptor II in renal fibrosis and inflammation in vivo and in vitro. J Pathol
227: 175-188, 2012.
60.
Meng XM, Huang XR, Xiao J, Chung AC, Qin W, Chen HY, Lan HY. Disruption of
Smad4 impairs TGF-beta/Smad3 and Smad7 transcriptional regulation during renal
inflammation and fibrosis in vivo and in vitro. Kidney Int 81: 266-279, 2012.
61.
Mezzano SA, Ruiz-Ortega M, Egido J. Angiotensin II and renal fibrosis. Hypertension
38: 635-638, 2001.
62.
Miyajima A, Chen J, Lawrence C, Ledbetter S, Soslow RA, Stern J, Jha S, Pigato J,
Lemer ML, Poppas DP, Vaughan ED, Felsen D. Antibody to transforming growth factor-beta
ameliorates tubular apoptosis in unilateral ureteral obstruction. Kidney Int 58: 2301-2313, 2000.
63.
Morath C, Dechow C, Lehrke I, Haxsen V, Waldherr R, Floege J, Ritz E, Wagner J.
Effects of retinoids on the TGF-beta system and extracellular matrix in experimental
glomerulonephritis. J Am Soc Nephrol 12: 2300-2309, 2001.
64.
Morrissey J, Hruska K, Guo G, Wang S, Chen Q, Klahr S. Bone morphogenetic
protein-7 improves renal fibrosis and accelerates the return of renal function. J Am Soc Nephrol
13 Suppl 1: S14-21, 2002.

25

657
658
659
660
661
662
663
664
665
666
667
668
669
670
671
672
673
674
675
676
677
678
679
680
681
682
683
684
685
686
687
688
689
690
691
692
693
694
695
696
697
698
699
700
701

65.
Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat
Rev Immunol 8: 958-969, 2008.
66.
Mucsi I, Skorecki KL, Goldberg HJ. Extracellular signal-regulated kinase and the
small GTP-binding protein, Rac, contribute to the effects of transforming growth factor-beta1 on
gene expression. J Biol Chem 271: 16567-16572, 1996.
67.
Neelisetty S, Alford C, Reynolds K, Woodbury L, Nlandu-Khodo S, Yang H, Fogo
AB, Hao CM, Harris RC, Zent R, Gewin L. Renal fibrosis is not reduced by blocking
transforming growth factor-beta signaling in matrix-producing interstitial cells. Kidney Int 88:
503-514, 2015.
68.
Ohashi K, Nagata K, Maekawa M, Ishizaki T, Narumiya S, Mizuno K. Rhoassociated kinase ROCK activates LIM-kinase 1 by phosphorylation at threonine 508 within the
activation loop. J Biol Chem 275: 3577-3582, 2000.
69.
Ono K, Ohtomo T, Ninomiya-Tsuji J, Tsuchiya M. A dominant negative TAK1
inhibits cellular fibrotic responses induced by TGF-beta. Biochem Biophys Res Commun 307:
332-337, 2003.
70.
Pallet N, Bouvier N, Legendre C, Gilleron J, Codogno P, Beaune P, Thervet E,
Anglicheau D. Autophagy protects renal tubular cells against cyclosporine toxicity. Autophagy
4: 783-791, 2008.
71.
Patel SR, Dressler GR. BMP7 signaling in renal development and disease. Trends Mol
Med 11: 512-518, 2005.
72.
Prelog M, Scheidegger P, Peter S, Gershwin ME, Wick G, Sgonc R. Diminished
transforming growth factor beta2 production leads to increased expression of a profibrotic
procollagen alpha2 type I messenger RNA variant in embryonic fibroblasts of UCD-200
chickens, a model for systemic sclerosis. Arthritis Rheum 52: 1804-1811, 2005.
73.
Qin W, Chung AC, Huang XR, Meng XM, Hui DS, Yu CM, Sung JJ, Lan HY. TGFbeta/Smad3 signaling promotes renal fibrosis by inhibiting miR-29. J Am Soc Nephrol 22: 14621474, 2011.
74.
RamachandraRao SP, Zhu Y, Ravasi T, McGowan TA, Toh I, Dunn SR, Okada S,
Shaw MA, Sharma K. Pirfenidone is renoprotective in diabetic kidney disease. J Am Soc
Nephrol 20: 1765-1775, 2009.
75.
Ren S, Babelova A, Moreth K, Xin C, Eberhardt W, Doller A, Pavenstadt H,
Schaefer L, Pfeilschifter J, Huwiler A. Transforming growth factor-beta2 upregulates
sphingosine kinase-1 activity, which in turn attenuates the fibrotic response to TGF-beta2 by
impeding CTGF expression. Kidney Int 76: 857-867, 2009.
76.
Robertson IB, Rifkin DB. Unchaining the beast; insights from structural and
evolutionary studies on TGFbeta secretion, sequestration, and activation. Cytokine Growth
Factor Rev 24: 355-372, 2013.
77.
Sato M, Muragaki Y, Saika S, Roberts AB, Ooshima A. Targeted disruption of TGFbeta1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral
ureteral obstruction. J Clin Invest 112: 1486-1494, 2003.
78.
Schaefer CJ, Ruhrmund DW, Pan L, Seiwert SD, Kossen K. Antifibrotic activities of
pirfenidone in animal models. Eur Respir Rev 20: 85-97, 2011.
79.
Sharma K, Ix JH, Mathew AV, Cho M, Pflueger A, Dunn SR, Francos B, Sharma S,
Falkner B, McGowan TA, Donohue M, Ramachandrarao S, Xu R, Fervenza FC, Kopp JB.
Pirfenidone for diabetic nephropathy. J Am Soc Nephrol 22: 1144-1151, 2011.

26

702
703
704
705
706
707
708
709
710
711
712
713
714
715
716
717
718
719
720
721
722
723
724
725
726
727
728
729
730
731
732
733
734
735
736
737
738
739
740
741
742
743
744
745
746

80.
Sharma K, Jin Y, Guo J, Ziyadeh FN. Neutralization of TGF-beta by anti-TGF-beta
antibody attenuates kidney hypertrophy and the enhanced extracellular matrix gene expression in
STZ-induced diabetic mice. Diabetes 45: 522-530, 1996.
81.
Shi M, Zhu J, Wang R, Chen X, Mi L, Walz T, Springer TA. Latent TGF-beta
structure and activation. Nature 474: 343-349, 2011.
82.
Shimizu T, Kuroda T, Hata S, Fukagawa M, Margolin SB, Kurokawa K. Pirfenidone
improves renal function and fibrosis in the post-obstructed kidney. Kidney Int 54: 99-109, 1998.
83.
Shull MM, Ormsby I, Kier AB, Pawlowski S, Diebold RJ, Yin M, Allen R, Sidman
C, Proetzel G, Calvin D, Annunziata N, Doetschman T. Targeted disruption of the mouse
transforming growth factor-beta 1 gene results in multifocal inflammatory disease. Nature 359:
693-699, 1992.
84.
Sugimoto H, Grahovac G, Zeisberg M, Kalluri R. Renal fibrosis and
glomerulosclerosis in a new mouse model of diabetic nephropathy and its regression by bone
morphogenic protein-7 and advanced glycation end product inhibitors. Diabetes 56: 1825-1833,
2007.
85.
Sureshbabu A, Bhandari V. Targeting mitochondrial dysfunction in lung diseases:
emphasis on mitophagy. Front Physiol 4: 384, 2013.
86.
Sureshbabu A, Ryter SW, Choi ME. Oxidative stress and autophagy: crucial
modulators of kidney injury. Redox Biol 4: 208-214, 2015.
87.
Sureshbabu A, Tonner E, Allan GJ, Flint DJ. Relative Roles of TGF-beta and IGFBP5 in Idiopathic Pulmonary Fibrosis. Pulm Med 2011: 517687, 2011.
88.
Suzuki HI, Kiyono K, Miyazono K. Regulation of autophagy by transforming growth
factor-beta (TGF-beta) signaling. Autophagy 6: 645-647, 2010.
89.
Trachtman H, Fervenza FC, Gipson DS, Heering P, Jayne DR, Peters H, Rota S,
Remuzzi G, Rump LC, Sellin LK, Heaton JP, Streisand JB, Hard ML, Ledbetter SR,
Vincenti F. A phase 1, single-dose study of fresolimumab, an anti-TGF-beta antibody, in
treatment-resistant primary focal segmental glomerulosclerosis. Kidney Int 79: 1236-1243, 2011.
90.
Tsou PS, Haak AJ, Khanna D, Neubig RR. Cellular mechanisms of tissue fibrosis. 8.
Current and future drug targets in fibrosis: focus on Rho GTPase-regulated gene transcription.
Am J Physiol Cell Physiol 307: C2-13, 2014.
91.
Wang B, Koh P, Winbanks C, Coughlan MT, McClelland A, Watson A, JandeleitDahm K, Burns WC, Thomas MC, Cooper ME, Kantharidis P. miR-200a Prevents renal
fibrogenesis through repression of TGF-beta2 expression. Diabetes 60: 280-287, 2011.
92.
Wang S, Chen Q, Simon TC, Strebeck F, Chaudhary L, Morrissey J, Liapis H,
Klahr S, Hruska KA. Bone morphogenic protein-7 (BMP-7), a novel therapy for diabetic
nephropathy. Kidney Int 63: 2037-2049, 2003.
93.
Wang S, Hirschberg R. Bone morphogenetic protein-7 signals opposing transforming
growth factor beta in mesangial cells. J Biol Chem 279: 23200-23206, 2004.
94.
Wang W, Huang XR, Li AG, Liu F, Li JH, Truong LD, Wang XJ, Lan HY.
Signaling mechanism of TGF-beta1 in prevention of renal inflammation: role of Smad7. J Am
Soc Nephrol 16: 1371-1383, 2005.
95.
Wilkes MC, Mitchell H, Penheiter SG, Dore JJ, Suzuki K, Edens M, Sharma DK,
Pagano RE, Leof EB. Transforming growth factor-beta activation of phosphatidylinositol 3kinase is independent of Smad2 and Smad3 and regulates fibroblast responses via p21-activated
kinase-2. Cancer Res 65: 10431-10440, 2005.

27

747
748
749
750
751
752
753
754
755
756
757
758
759
760
761
762
763
764
765
766

96.
Xu L, Xiong S, Guo R, Yang Z, Wang Q, Xiao F, Wang H, Pan X, Zhu M.
Transforming growth factor beta3 attenuates the development of radiation-induced pulmonary
fibrosis in mice by decreasing fibrocyte recruitment and regulating IFN-gamma/IL-4 balance.
Immunol Lett 162: 27-33, 2014.
97.
Xu Y, Yang S, Huang J, Ruan S, Zheng Z, Lin J. Tgf-beta1 induces autophagy and
promotes apoptosis in renal tubular epithelial cells. Int J Mol Med 29: 781-790, 2012.
98.
Yi JY, Shin I, Arteaga CL. Type I transforming growth factor beta receptor binds to and
activates phosphatidylinositol 3-kinase. J Biol Chem 280: 10870-10876, 2005.
99.
Yu L, Border WA, Huang Y, Noble NA. TGF-beta isoforms in renal fibrogenesis.
Kidney Int 64: 844-856, 2003.
100. Zeisberg M, Hanai J, Sugimoto H, Mammoto T, Charytan D, Strutz F, Kalluri R.
BMP-7 counteracts TGF-beta1-induced epithelial-to-mesenchymal transition and reverses
chronic renal injury. Nat Med 9: 964-968, 2003.
101. Zhou TB, Drummen GP, Qin YH. The controversial role of retinoic acid in fibrotic
diseases: analysis of involved signaling pathways. Int J Mol Sci 14: 226-243, 2012.
102. Ziyadeh FN, Hoffman BB, Han DC, Iglesias-De La Cruz MC, Hong SW, Isono M,
Chen S, McGowan TA, Sharma K. Long-term prevention of renal insufficiency, excess matrix
gene expression, and glomerular mesangial matrix expansion by treatment with monoclonal
antitransforming growth factor-beta antibody in db/db diabetic mice. Proc Natl Acad Sci U S A
97: 8015-8020, 2000.

767
768

28

769

FIGURE LEGENDS

770

Figure 1. Smad and non-Smad TGF-1 signaling pathways. (A) TGF-1 activation occurs

771

with the release from latent TGF- binding protein (LTBP) complex by proteases. TGF-1

772

signaling is initiated upon binding of active TGF-1 with TRII and forming TRI-TRII

773

heteromeric complex, leading to phosphorylation of Smad2/3, oligomerization with Smad4, and

774

subsequent nuclear translocation to regulate the transcription of ECM genes. Smad7 serves as a

775

negative regulator of TGF-1 signaling. TGF-1 also activates Smad-independent signaling such

776

as the MAPK, mediated by Ras-Raf-MEK-ERK, pathway, and TGF--activated kinase 1

777

(TAK1) mediated by TAK1- TAK1-binding protein 1 (TAB1) pathway. TGF-1-induced TAK1

778

activation leads to signaling via MKK4-JNK and MKK3-p38 pathways and activation of

779

transcription factors activator protein-1 (AP-1) and activating transcription factor 2 (ATF-2),

780

respectively, and activation of NF-B to mediate pro-fibrotic responses.

781
782

Figure 2. An overview of Smad signaling and inflammatory pathways of TGF-1 signaling.

783

Binding of Smad4 to phosphorylated Smad2/3 leads to the nuclear translocation of Smad

784

complex to regulate gene transcription including Smad7. Smad7 induces IB expression which

785

inhibits phosphorylation of p65/p50 proteins of NFB signaling complex to prevent NFB-

786

driven renal inflammation.

787
788

Figure 3. An overview of pro-fibrotic and protective pathways of TGF-1 signaling.

789

Activation of TGF-1 in response to kidney injury can signal both pro-fibrotic and protective

790

effects via Smad and non-Smad signaling pathways. Overexpression of Smad2 attenuates TGF-

29

791

1-induced Smad3 phosphorylation and type I collagen (Col-11) expression, whereas inhibition

792

of Smad2 promotes renal fibrosis via enhanced TGF-1/Smad3 signaling. TGF-1-induced

793

autophagy negatively regulates TGF-1-stimulated collagen accumulation in the kidney by

794

promoting collagen degradation. Autophagy also negatively regulates mature TGF-1

795

expression, thereby protects against kidney fibrosis.

30

Table 1. Protective roles of TGF- signaling in chronic kidney diseases.


Strategy and Results
Conditional deletion of TRII in renal interstitial cells
significantly reduces collagen production, but does not
ameliorate overall renal fibrosis

Disease Model
UUO,
Aristolochic acidinduced
nephropathy
UUO

(58)

Overexpression of latent TGF-1 in keratinocytes reduces renal


fibrosis

UUO

(28)

Overexpression of latent TGF-1 in keratinocytes protects


against glomerular crescentic formation and severe
tubulointerstitial damage
Overexpression of Smad7 suppresses renal fibrosis, whereas
deficiency of Smad7 aggravates the severity of renal fibrosis

Crescentic
glomerulonephritis

(29)

Diabetic
nephropathy

(8)

Smad7 overexpression in kidneys suppresses renal fibrosis and


disruption of Smad7 enhances renal fibrosis

UUO

(14, 46)

Smad 7 deficiency aggravates angiotensin II-mediated renal


fibrosis, whereas Smad7 treatment prevents progressive renal
injury

Hypertensive
nephropathy

(51, 52)

Smad7 disruption exacerbates nephropathy, whereas Smad7


overexpression in kidneys attenuates nephropathy

Aristolochic acidinduced
nephropathy

(15)

Conditional deletion of TRII in renal tubular epithelial cells


and renal fibroblasts enhances NF-B signaling and renal
inflammation

UUO

(59)

Conditional deletion of Smad4 in renal tubular epithelial cells


enhances renal inflammation

UUO

(60)

Treatment with autophagy inhibitor 3-Methyladenine enhances


renal tubular cell apoptosis and tubulointerstitial fibrosis

UUO

(41)

Heterozygous deletion of Beclin 1 and LC3b-null mice


enhances collagen deposition

UUO

(19, 40)

Conditional deletion of Atg5 in podocytes increases


susceptibility to renal injury

Puromycin,
Adriamycin,
Lipopolysaccharide

(27)

Conditional deletion of Smad2 in renal tubular epithelial cells


enhances renal fibrosis

References
(67)

Table 2. Recent clinical trials using anti-TGF- therapy in fibrotic kidney diseases.

Compound
LY2382770

Target
TGF-1

Company
Eli Lilly

Fresolimumab TGF-1,2,3 Genzyme


(GC1008)

Pirfenidone

TGF-1,2,3 InterMune

Phase

Disease

Phase II

Diabetic
nephropathy

Phase I

Focal Segmental
Glomerulosclerosis

References or
NCT
Identifier
NCT01113801
(89)

Phase II

NCT01665391

Phase II

Focal Segmental
NCT00001959
Glomerulosclerosis, (11)

Phase I/II

Diabetic
nephropathy

NCT00063583
(79)

Figure 2

Figure 3

You might also like