Abstract
Purpose of review:
Pre-clinical data suggests that transforming growth factor-β (TGF-β) is arguably the most potent profibrotic growth factor in kidney injury. Despite this, recent clinical trials targeting TGF-β have been disappointing. These negative studies suggest that TGF-β signaling in the injured kidney might be more complicated than originally thought. This review examines recent studies that expand our understanding of how this pleiotropic growth factor affects renal injury.
Recent findings:
There are recent studies showing new mechanisms whereby TGF-β can mediate injury (e.g. epigenetic effects, macrophage chemoattractant). However, more significant are the increasing reports on cross-talk between TGF-β signaling and other pathways relevant to renal injury such as Wnt/β-catenin, YAP/TAZ, and klotho/FGF23. TGF-β clearly alters the response to injury, not just by direct transcriptional changes on target cells, but also through effects on other signaling pathways. In T cells and tubular epithelial cells, some of these TGF-β-mediated changes are potentially beneficial.
Summary:
It is unlikely that inhibition of TGF-β per se will be a successful antifibrotic strategy, but a better understanding of TGF-β’s actions may reveal promising downstream targets or modulators of signaling to target therapeutically for chronic kidney disease.
Keywords: renal injury, fibrosis, growth factors
Introduction
Transforming growth factor-β (TGF-β is a pleiotropic growth factor that plays important roles in organ development, immune responses, cancer biology, and the response to injury. In the kidney, TGF-β is considered a key mediator of fibrosis, the accumulation of extracellular matrix (ECM), in both the glomerulus (glomerulosclerosis) and the tubulointerstitial compartment (tubulointerstitial fibrosis) after injury. Tubulointerstitial fibrosis progression is a common pathologic feature of all renal injuries (e.g. hypertension, diabetes, glomerulonephritides) that lead to loss of renal function and eventual end-stage renal disease. Recent studies confirm the importance of tubulointerstitial fibrosis on renal biopsies as a prognostic indicator of renal survival even in glomerular injuries(1, 2). Given the rise in chronic kidney disease globally, TGF-β has received much attention as a potential anti-fibrotic target.
There are three mammalian isoforms of TGF-β (−β1, −β2, −β3) that are part of the TGF-β superfamily which also include bone morphogenic proteins and activins. The TGF-β ligands are secreted as inactive proteins due to noncovalent binding to the latency-associated peptide (LAP), and this complex is tethered to the ECM by the latent TGF-β binding proteins(3). TGF-β is activated through proteolytic cleavage from the LAP by several proteases such as matrix metalloproteinases (MMPs) and plasmin(4). In addition, integrins containing αv can bind to RGD domains of the LAP and facilitate mechanical release and activation(5). Augmented mechanical tension as well as increased expression of integrins and MMPs are induced in the fibrotic kidney, leading to many potential activators of TGF-β in injury. Active TGF-β ligands all bind the TGF-β type II receptor (TβRII) which then heterodimerizes to the TGF-β type I receptor (TβRI)(6). The activated TβRI (also called ALK5) phosphorylates Smad2 and 3 which bind to the common Smad4 and accumulate in the nucleus to alter gene expression(6, 7). In addition to activating Smad-dependent (i.e. canonical) transcriptional responses, the activated receptor complex activates many other signaling proteins such as mitogen-activated protein kinase (MAPK), AKT, and Rho GTPases(8). The diversity of signaling pathways downstream of the activated TβRII/TβRI complex likely accounts for its pleiotropic effects that vary based upon target cell type and microenvironment.
There is abundant evidence that TGF-β promotes renal fibrosis from rodent models in which TGF-β activity has been systemically modulated. Transgenic mice with increased circulating TGF-β developed glomerulosclerosis and those with increased tubular production of TGF-β sustained tubulointerstitial fibrosis in the absence of any additional injury(9, 10). Conversely, systemically inhibiting TGF-β ameliorated tubulointerstitial fibrosis and glomerulosclerosis in the unilateral ureteral obstruction (UUO) and glomerulonephritis models, respectively(11, 12). Genetic inhibition of Smad3, but not Smad2, reduced ECM after injury by UUO, supporting a role for TGF-β/Smad3 signaling in renal fibrosis(13, 14). Most cells express the TGF-β receptor complex, so fully understanding how TGF-β signaling in diverse cell populations alters the response to injury has been challenging.
Initial studies investigating TGF-β and renal fibrosis focused on this growth factor’s effects on fibroblasts. TGF-β promotes activation of fibroblasts to myofibroblasts with increased α-smooth muscle actin (α-SMA) expression, synthesis of ECM components, and fibroblast proliferation(15–17). Myofibroblasts, potent producers of ECM, are widely considered the main cell type responsible for fibrosis progression. There is also data that TGF-β sensitizes epithelial cells to apoptosis and tubular degeneration leading to tubular atrophy, a component of tubulointerstitial fibrosis(10, 11). Despite the detrimental effects of TGF-β in murine models of CKD, recent clinical trials did not show a benefit in blocking TGF-β for diabetic nephropathy or focal segmental glomerulosclerosis(18, 19). There are many potential explanations for these negative studies, but it raises the possibility that TGF-β signaling in the injured kidney may not be completely understood. This review highlights some of the newer data involving TGF-β and the kidney including potentially beneficial effects of this Jekyll-and-Hyde protein (Table 1) and new interactions between TGF-β and other growth factors/signaling pathways (Figure 1). The newer data still point to an important profibrotic role for TGF-β and renal injury but paint a more nuanced and complicated picture of how this multi-faceted growth factor affects the response to injury.
Table 1.
TGF-β-dependent effects in renal injury
| TGF-β-related Effect | Injury/Model | Effect on Renal Injury | Ref. |
|---|---|---|---|
| TGF- β acts as chemoattractantfor macrophages | Deleted TβRII in macrophages (CDIIb-Cre and LysM-Cre) I/R model of AKI and AKI/CKD and HB-EGF transgenic models | Reduced inflammation and fibrosis | 28 |
| TGF- β promotes regulatory T cells (Treg) differentiation | Exogenous TGF- β plus inhibitor of β -catenin/TCF I/R and UUO models β | Reduced fibrosis | 37 |
| Genetic inhibition of TGF- β signaling in tubules increased apoptosis and G2 arrest after injury | TβRII deleted in proximal tubules (γGT-Cre) Aristolochic acid injury and Uninephrectomy with angiotensin II | Fibrosis increased and renal function impaired | 43 |
| TGF- β inhibits klotho through methylation | Pharmacologic and genetic inhibition of TGF-β-dependent methyltransferase UUO model | Reduced fibrosis | 50, 51 |
| TGF- β promotes profibrotic gene transcription through SET7/9-dependent methylation of H3K4 | siRNA to SET7/9 or sinefungin (SET7/9 inhibitor) UUO model and peritoneal fibrosis murine models | Reduced fibrosis and α-SMA expression | 54,55 |
| FGF23 stimulated TGF- β and JNK | Fibroblasts isolated from UUO kidneys | Increased myofibroblast differentiation and ECM production | 56 |
| TGF- β stimulates TAZ expression | C3H/10T1/2 cells (mouse mesenchymal cells) and LLC-PK1 (porcine tubule epithelial) or HK-2 cells* | *Epithelial de-differentiation, vimentin expression, cell cycle arrest | 59, 60* |
| TGF- β signaling suppresses angiogenesis | Heterozygous TβRII deletion in endothelial cells (TβRMflox/+Tie2-Cre) Folic acid nephropathy and UUO | Protected against fibrosis | 63 |
| TGF- β stimulates epithelial cells and macrophages to produce VEGF-C | Induction of VEGF-C suppressed by TβRI/ALK5 inhibitor | Reduced lymphangiogenesis | 69 |
| CTGF as mediator of TGF- β -dependent lymphangiogenesis | CTGF knockout mice UUO model | Reduced VEGF-C and lymphangiogenesis | 70 |
This table lists recently described TGF-β-dependent effects, the model in which these effects were observed, their impact on renal injury, and the corresponding reference(s).
Figure 1. TGF-β interactions with other signaling pathways in renal injury.
Many effects of TGF-β on renal injury depend upon interactions with other growth factor pathways. This signaling cross-talk as well as the target cell types in which these interactions have been described are shown. TGF-β had protective effects on T cells and tubule epithelial cells through modulation of β-catenin signaling(37, 41). TGF-β increases expression of the Hippo transcription factor TAZ(59, 60). The protective klotho, a receptor for FGF23, is suppressed by TGF-β-dependent methylation in tubule epithelial cells(50, 51), and some profibrotic effects of FGF23 in fibroblasts are mediated by downstream TGF-β signaling(56, 57). TGF-β stimulated tubular epithelial cells and macrophages to produce VEGF-C, a potent inducer of lymphangiogenesis, and some of these effects may be mediated by CTGF(69, 70).
TGF-β, transforming growth factor-β; FGF23, fibroblast growth factor 23; VEGF-C, vascular endothelial growth factor-C; CTGF, connective tissue growth factor.
TGF-β and Renal Inflammation
In homeostasis, TGF-β has well-defined anti-inflammatory effects, and mice lacking TGF-β1 or TβRII die soon after birth with overwhelming inflammation(20, 21). The role of TGF-β on inflammation after renal injury is more complex. Macrophages play key roles in the response to acute kidney injury (AKI) with proinflammatory M1 (classically activated) macrophages infiltrating early in animal models of AKI. Reducing macrophage infiltration at this early stage has been shown to be beneficial(22). At later stages following AKI, a reparative M2 (alternatively activated) macrophage subtype predominates, and macrophage depletion can impair recovery and promote chronic kidney disease (CKD)(23). The role of macrophages in CKD is less clear, but macrophage infiltration on human CKD biopsies predicts worse renal outcomes(24). TGF-β signaling can induce a M2 macrophage polarization which may suppress inflammation but, if persistent, can also promote fibrosis through the production of pro-fibrotic growth factors such as TGF-β1(25, 26). However, selective deletion of TGF-β1 from macrophages did not reduce fibrosis after murine models of injury, suggesting that other macrophage-derived growth factors may be mediating fibrosis(27). A recent study deleted TβRII in macrophages and showed reduced tubulointerstitial fibrosis and macrophage infiltration following injury(28). TGF-β-infused Matrigel plugs in vivo acted as a powerful chemoattractant for wildtype but not TβRII−/− macrophages(28). Furthermore, fewer labeled bone-marrow derived TβRII−/− monocytes migrated to the injured kidney compared to WT monocytes after an ischemic AKI/CKD model(28). Interestingly, the conditional deletion of TβRII didn’t alter the ratio of M1 and M2 macrophages. These studies indicate that TGF-β promotes fibrosis, in part, through its role as a macrophage chemoattractant, but macrophages likely promote fibrosis through production of cytokines other than TGF-β1.
T cells are another inflammatory cell type with divergent effects depending upon the microenvironment. TGF-β can promote an anti-inflammatory, Foxp3+, regulatory T cell subset (Treg) which enhances repair after ischemic injury(29, 30). However, in the presence of IL-6, TGF-β induces T cell differentiation to a more inflammatory T17 subtype(31). Several studies describe both pro- and anti-inflammatory effects of TGF-β after renal injury depending upon the type of injury and method of altering TGF-β signaling(32–35). A recent study showed that exogenous TGF-β may reduce inflammation and fibrosis through effects on the Wnt/β-catenin pathway. The transcription factor β-catenin, the canonical signaling protein of Wnt, usually binds to LEF/TCF transcription factors. The transcription factor FoxO is known to compete with LEF/TCF for binding to β-catenin(36), and TGF-β increased β-catenin/FoxO1 interactions(37). When coupled with an inhibitor of β-catenin/TCF interactions (ICG-001), exogenous TGF-β1 reduced kidney fibrosis through the induction of Tregs(37). This effect was abrogated by the depletion of Tregs in both the ischemia/reperfusion (I/R) and UUO models. This study exemplifies how TGF-β can mediate opposing effects on injury response based upon its interactions with other growth factor pathways and signaling proteins.
TGF-β and Epithelial Cells
Increased TGF-β levels impair the epithelial response to injury by sensitizing renal tubules and podocytes to apoptosis and enhancing de-differentiation(11, 38). Epithelial de-differentiation may compromise tubule transport/function and can lead to increased production of profibrotic and pro-inflammatory cytokines(39, 40). More recent studies have expanded upon the cellular effects TGF-β exerts on epithelia. Similar to the protective TGF-β/β-catenin interactions noted above with T cells, our group reported that selective deletion of proximal tubular TβRII worsened tubular apoptosis and fibrosis in murine CKD models, in part, through reduced β-catenin activity(41). Excessive TGF-β signaling is clearly deleterious to injured tubules, but TGF-β mediates many diverse cellular responses and its inhibition may cause loss of some protective effects. Others have also reported potentially beneficial effects of TGF-β through its effects on autophagy(42). Our group found that proximal tubule cells lacking TβRII had increased G2/M arrest, associated with a profibrotic phenotype(43), after aristolochic acid exposure in vitro(41). Interestingly, treatment of wild type cells with TGF-β1 (10ng/mL but not 1ng/mL) also induced G2/M arrest (unpublished data). TGF-β signaling can affect many different stages of the cell cycle (e.g. p15, p21), and the effect likely varies depending upon the dose and target cell type. One limitation is the ability to measure active TGF-β in vivo to know what doses in vitro correlate with renal injury. These divergent, dose-dependent effects of TGF-β have also been found in studies looking at TGF-β-dependent matrix production and inflammation(44, 45). How epithelial TGF-β signaling alters the cell cycle in renal injury and how these cell cycle changes affect tubular atrophy and tubulointerstitial fibrosis requires further investigation.
In addition to the β-catenin pathway, TGF-β signaling also affects expression of klotho, an essential subunit of the fibroblast growth factor-23 (FGF-23) receptor(46). Klotho is produced in renal tubule cells and is an important suppressor of aging, but its expression is reduced after renal injury and in human CKD(47–49). Recent data suggests that epithelial TGF-β signaling suppresses klotho expression through methylation. One group has shown that TGF-β mediates this response by induction of a methyltransferase (G9a) that targets histone H3 at a locus known to regulate klotho gene expression in cancer cells(50). Another group has shown that TGF-β alters methylation of the klotho promoter indirectly through miRNAs that inhibit methyltransferases(51). These recent studies suggest that TGF-β-dependent suppression of klotho may be another mechanism by which excessive TGF-β is deleterious to injured tubules.
TGF-β and Mesenchymal Cells
As mentioned earlier, TGF-β stimulates fibrosis by inducing fibroblasts to transform into myofibroblasts, increase ECM production, and augment proliferation, migration, and adhesion. These TGF-β-dependent effects also occur with pericytes, a recently recognized potent source of myofibroblasts after injury(52, 53). More recent data add to our understanding of TGF-β and mesenchymal cells primarily by defining new modulators of TGF-β signaling and new interactions with other important signaling pathways. TGF-β-dependent epigenetic changes in epithelial cells were mentioned above, and many studies have identified a similar mechanism whereby TGF-β1 alters fibroblast protein expression. TGF-β1 induced methylation of lysine 4 of histone H3 (H3K4) by upregulation of SET-domain containing lysine methyltransferase 7/9 (SET7/9) in both renal fibroblasts and epithelial cells(54). Treating obstructed mice with siRNA to SET7/9 reduced renal fibrosis. Moreover, an inhibitor of SET7/9, sinefungin, inhibited H3K4 methylation and reduced TGF-β1-dependent α-SMA expression in both renal epithelial cells and fibroblasts(54). TGF-β1 upregulated SET7/9 in a Smad3-dependent manner, and TGF-β1 increased mono-methylated H3K4 levels in the promoters of Col1a1, CTGF, PAI-1. The same group also reported an increase in SET7/9 expression in peritoneal fluid from peritoneal dialysis patients (compared to control human peritoneal mesothelial cells), and sinefungin reduced peritoneal fibrosis in murine models(55). These data suggest that altered methylation is another mechanism by which TGF-β1 stimulates profibrotic gene expression in mesenchymal cells.
Recent studies have also shed light on TGF-β interactions with the FGF23/klotho pathway in fibroblasts isolated from UUO-injured kidneys (UUOF). FGF23 stimulation of UUOF led to TGF-β-dependent JNK signaling(56). FGF23 signaled through the FGFR4 receptor and induced increased intracellular calcium via the channel TRPC6. This led to augmented reactive oxygen species (ROS) and TGF-β-dependent JNK activation(56). Smad-dependent TGF-β effects downstream of FGF23 have also been shown(57). The effects in both studies were present in UUOF but not normal (i.e. uninjured) fibroblasts, underscoring the limitations with using NRK-49F cells. These studies were performed in vitro and need to be assessed in the injured kidney, but they expand the known interactions between TGF-β and FGF/klotho pathways.
The Hippo pathway transcriptional co-activators YAP and TAZ also have important cross-talk with TGF-β signaling. Nuclear YAP and TAZ interact with Smads2/3 and promote Smad activity through nuclear retention, but cytosolic YAP and TAZ inhibit Smad activation. The localization of YAP/TAZ is altered by substrate stiffness and cell density, leading to increased TGF-β signaling with reduced cell density or increased matrix stiffness(58). Conversely, it was recently shown that TGF-β signaling can increase TAZ expression in both epithelial and mesenchymal cells from the kidney by two independent groups(59, 60). TGF-β increased TAZ transcription through p38-dependent signaling but did not alter TAZ nuclear localization(59). By increasing the total amount of TAZ (but not YAP), TGF-β signaling could augment TAZ signaling in the presence of an activator. In the context of renal injury, it is likely that TGF-β and YAP/TAZ act in a bi-directional, synergistic manner to promote fibrosis.
TGF-β and Vasculature
The renal microvasculature, including peritubular capillary endothelial cells, pericytes, and surrounding lymphatics, is emerging as an important component of the renal response to injury. The importance of TGF-β signaling on pericytes is well-documented and discussed under Mesenchymal Cells. A decrease in capillary density was initially noted one month after rats underwent the I/R ischemic injury, and these vascular defects persisted for 10 months after the acute injury concurrent with tubulointerstitial fibrosis(61). These results imply that the vascular defects from severe AKI play a pathophysiologic role in the subsequent development of fibrosis and CKD. The role of TGF-β signaling in the injured vasculature is complicated as TGF-β can promote expression of both the pro-angiogenic factor VEGF-A and the anti-angiogenic factor thrombospondin-1(62). More recent data suggest that excessive TGF-β signaling is deleterious to the injured endothelium. Heterozygous deletion of TβRII in the endothelium (TβRIIflox/+;Tie2-Cre) was protective against fibrosis in both the folic acid nephropathy and UUO models(63). In the endothelial cells specifically, TGF-β can also activate the ALK1 receptor (rather than ALK5/ TβRI) which leads to Smads1/5 activation and pro-angiogenic signaling(63). Heterozygous knockout of TβRII in endothelial cells resulted in more proangiogenic Smad1/5 and less fibrotic Smad2/3 activation(63).
The lymphatic vessels, lined by lymphatic endothelial cells (LECs), undergo new growth, or lymphangiogenesis, during injury, but its role in renal injury and fibrosis remains unclear. The lymphatics drain interstitial fluid and return it to the intravascular space and are also involved in the trafficking of inflammatory cells(64). Lymphangiogenesis occurs in murine models of renal and peritoneal fibrosis, and the increase in lymphatic vessels in human diabetic nephropathy biopsies correlated with both the extent of inflammation and fibrosis(65). There are conflicting reports about whether TGF-β signaling stimulates or suppresses lymphangiogenesis, but most agree that the growth factor VEGF-C promotes growth of new lymphatic vessels(66). There are many potential explanations for the diverse effects of TGF-β on lymphatics, but most of the inhibitory studies examined the direct role of TGF-β on LECs in vitro(67, 68). By contrast, others found that TGF-β stimulated tubule epithelial cells and macrophages to produce VEGF-C, leading indirectly to lymphangiogenesis(69). More recently, a group found that connective tissue growth factor (CTGF) may be a downstream mediator of TGF-β-dependent lymphangiogenesis in renal injury(70). Lymphangiogenesis is likely an important modulator of the inflammatory response, and more studies are necessary to determine how TGF-β-dependent changes in lymph vessel growth and function impact the progression of renal fibrosis.
Conclusion
TGF-β affects many diverse cells’ responses to renal injury (Table 1). Although excessive TGF-β signaling is clearly profibrotic, this growth factor mediates some protective effects on inflammation and epithelial cell function depending upon the microenvironment. Recent data also point to the importance of cross-talk between TGF-β and other signaling pathways in the development of renal fibrosis.
Key Points:
TGF-β is a pleiotropic growth factor that mediates different cellular responses depending upon the target cell type and microenvironment.
Many of TGF-β’s effects are likely due to interactions with other growth factor signaling pathways (Figure 1).
Some TGF-β signaling in T cells and tubular epithelia may exert protective effects on injury through β-catenin signaling.
Acknowledgements:
Funding for the author was provided by NIDDK R01DK108968–01 and VA Merit 1I01BX003425-01A1. There are no conflicts of interest to disclose.
Disclosure of Funding:
National Institutes of Health: NIDDK R01DK108968–01
Veterans Affairs: VA Merit 1I01BX003425–01A1
Reference section
- 1.Mariani LH, Martini S, Barisoni L, et al. Interstitial fibrosis scored on whole-slide digital imaging of kidney biopsies is a predictor of outcome in proteinuric glomerulopathies. Nephrol Dial Transplant 2018;33(2):310–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Nath KA. Tubulointerstitial changes as a major determinant in the progression of renal damage. Am J Kidney Dis 1992;20(1):1–17. [DOI] [PubMed] [Google Scholar]
- 3.Rifkin DB. Latent transforming growth factor-beta (TGF-beta) binding proteins: orchestrators of TGF-beta availability. J Biol Chem 2005;280(9):7409–12. [DOI] [PubMed] [Google Scholar]
- 4.Jenkins G The role of proteases in transforming growth factor-beta activation. Int J Biochem Cell Biol 2008;40(6–7):1068–78. [DOI] [PubMed] [Google Scholar]
- 5.Munger JS, Huang X, Kawakatsu H, et al. The integrin alpha v beta 6 binds and activates latent TGF beta 1: a mechanism for regulating pulmonary inflammation and fibrosis. Cell. 1999;96(3):319–28. [DOI] [PubMed] [Google Scholar]
- 6.Wrana JL, Attisano L, Carcamo J, et al. TGF beta signals through a heteromeric protein kinase receptor complex. Cell. 1992;71(6):1003–14. [DOI] [PubMed] [Google Scholar]
- 7.Brown KA, Pietenpol JA, Moses HL. A tale of two proteins: differential roles and regulation of Smad2 and Smad3 in TGF-beta signaling. J Cell Biochem 2007;101(1):9–33. [DOI] [PubMed] [Google Scholar]
- 8.Zhang YE. Non-Smad Signaling Pathways of the TGF-beta Family. Cold Spring Harb Perspect Biol 2017;9(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kopp JB, Factor VM, Mozes M, et al. Transgenic mice with increased plasma levels of TGF-beta 1 develop progressive renal disease. Laboratory investigation; a journal of technical methods and pathology. 1996;74(6):991–1003. [PubMed] [Google Scholar]
- 10.Koesters R, Kaissling B, Lehir M, et al. Tubular overexpression of transforming growth factor-beta1 induces autophagy and fibrosis but not mesenchymal transition of renal epithelial cells. Am J Pathol 2010;177(2):632–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Miyajima A, Chen J, Lawrence C, et al. Antibody to transforming growth factor-beta ameliorates tubular apoptosis in unilateral ureteral obstruction. Kidney Int 2000;58(6):2301–13. [DOI] [PubMed] [Google Scholar]
- 12.Border WA, Okuda S, Languino LR, et al. Suppression of experimental glomerulonephritis by antiserum against transforming growth factor beta 1. Nature. 1990;346(6282):371–4. [DOI] [PubMed] [Google Scholar]
- 13.Sato M, Muragaki Y, Saika S, et al. Targeted disruption of TGF-beta1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction. J Clin Invest 2003;112(10):1486–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Meng XM, Huang XR, Chung AC, et al. Smad2 protects against TGF-beta/Smad3-mediated renal fibrosis. J Am Soc Nephrol 2010;21(9):1477–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Desmouliere A, Geinoz A, Gabbiani F, et al. Transforming growth factor-beta 1 induces alpha-smooth muscle actin expression in granulation tissue myofibroblasts and in quiescent and growing cultured fibroblasts. J Cell Biol 1993;122(1):103–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ignotz RA, Endo T, Massague J. Regulation of fibronectin and type I collagen mRNA levels by transforming growth factor-beta. J Biol Chem 1987;262(14):6443–6. [PubMed] [Google Scholar]
- 17.Roberts AB, Sporn MB, Assoian RK, et al. Transforming growth factor type beta: rapid induction of fibrosis and angiogenesis in vivo and stimulation of collagen formation in vitro. Proc Natl Acad Sci U S A 1986;83(12):4167–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Voelker J, Berg PH, Sheetz M, et al. Anti-TGF-beta1 Antibody Therapy in Patients with Diabetic Nephropathy. J Am Soc Nephrol 2017;28(3):953–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Vincenti F, Fervenza FC, Campbell KN, et al. A Phase 2, Double-Blind, Placebo-Controlled, Randomized Study of Fresolimumab in Patients With Steroid-Resistant Primary Focal Segmental Glomerulosclerosis. Kidney Int Rep 2017;2(5):800–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Shull MM, Ormsby I, Kier AB, et al. Targeted disruption of the mouse transforming growth factor-beta 1 gene results in multifocal inflammatory disease. Nature. 1992;359(6397):693–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Leveen P, Larsson J, Ehinger M, et al. Induced disruption of the transforming growth factor beta type II receptor gene in mice causes a lethal inflammatory disorder that is transplantable. Blood. 2002;100(2):560–8. [DOI] [PubMed] [Google Scholar]
- 22.Day YJ, Huang L, Ye H, et al. Renal ischemia-reperfusion injury and adenosine 2A receptor-mediated tissue protection: role of macrophages. Am J Physiol Renal Physiol 2005;288(4):F722–31. [DOI] [PubMed] [Google Scholar]
- 23.Lee S, Huen S, Nishio H, et al. Distinct macrophage phenotypes contribute to kidney injury and repair. J Am Soc Nephrol 2011;22(2):317–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Eardley KS, Kubal C, Zehnder D, et al. The role of capillary density, macrophage infiltration and interstitial scarring in the pathogenesis of human chronic kidney disease. Kidney Int 2008;74(4):495–504. [DOI] [PubMed] [Google Scholar]
- 25.Zhang F, Wang H, Wang X, et al. TGF-beta induces M2-like macrophage polarization via SNAIL-mediated suppression of a pro-inflammatory phenotype. Oncotarget. 2016;7(32):52294–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol 2011;11(11):723–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.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 2013;305(4):F477–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Chung S, Overstreet JM, Li Y, et al. TGF-beta promotes fibrosis after severe acute kidney injury by enhancing renal macrophage infiltration. JCI Insight. 2018;3(21).*This recent study shows that TGF-b’s function as a macrophage chemoattractant increases fibrosis in a model of AKI to CKD. Macrophages lacking the TbRII receptor had reduced accumulation in the injured kidney though chemotaxis to other stimuli was not affected.
- 29.Gandolfo MT, Jang HR, Bagnasco SM, et al. Foxp3+ regulatory T cells participate in repair of ischemic acute kidney injury. Kidney Int 2009;76(7):717–29. [DOI] [PubMed] [Google Scholar]
- 30.Letterio JJ. TGF-beta signaling in T cells: roles in lymphoid and epithelial neoplasia. Oncogene. 2005;24(37):5701–12. [DOI] [PubMed] [Google Scholar]
- 31.Bettelli E, Carrier Y, Gao W, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441(7090):235–8. [DOI] [PubMed] [Google Scholar]
- 32.Nath KA, Croatt AJ, Warner GM, et al. Genetic deficiency of Smad3 protects against murine ischemic acute kidney injury. Am J Physiol Renal Physiol 2011;301(2):F436–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Prakash J, de Borst MH, van Loenen-Weemaes AM, et al. Cell-specific delivery of a transforming growth factor-beta type I receptor kinase inhibitor to proximal tubular cells for the treatment of renal fibrosis. Pharm Res 2008;25(10):2427–39. [DOI] [PubMed] [Google Scholar]
- 34.Wang W, Huang XR, Li AG, et al. Signaling mechanism of TGF-beta1 in prevention of renal inflammation: role of Smad7. J Am Soc Nephrol 2005;16(5):1371–83. [DOI] [PubMed] [Google Scholar]
- 35.Meng XM, Huang XR, Xiao J, et al. Disruption of Smad4 impairs TGF-beta/Smad3 and Smad7 transcriptional regulation during renal inflammation and fibrosis in vivo and in vitro. Kidney Int 2012;81(3):266–79. [DOI] [PubMed] [Google Scholar]
- 36.Almeida M, Han L, Martin-Millan M, et al. Oxidative stress antagonizes Wnt signaling in osteoblast precursors by diverting beta-catenin from T cell factor- to forkhead box O-mediated transcription. J Biol Chem 2007;282(37):27298–305. [DOI] [PubMed] [Google Scholar]
- 37.Qiao X, Rao P, Zhang Y, et al. Redirecting TGF-beta Signaling through the beta-Catenin/Foxo Complex Prevents Kidney Fibrosis. J Am Soc Nephrol 2018;29(2):557–70.** This study shows that TGF-b, in addition to an inhibitor of b-catenin/TCF interactions, can be protective in renal injury. The proposed mechanism is re-directed b-catenin signaling away from TCF and through the FoxO1 transcription factor. The combination of exogenous TGF-b plus enhanced b-catenin/FoxO1 signaling led to increased anti-inflammatory Treg differentiation.
- 38.Wu DT, Bitzer M, Ju W, et al. TGF-beta concentration specifies differential signaling profiles of growth arrest/differentiation and apoptosis in podocytes. J Am Soc Nephrol 2005;16(11):3211–21. [DOI] [PubMed] [Google Scholar]
- 39.Lovisa S, LeBleu VS, Tampe B, et al. Epithelial-to-mesenchymal transition induces cell cycle arrest and parenchymal damage in renal fibrosis. Nature medicine. 2015;21(9):998–1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Grande MT, Sanchez-Laorden B, Lopez-Blau C, et al. Snail1-induced partial epithelial-to-mesenchymal transition drives renal fibrosis in mice and can be targeted to reverse established disease. Nature medicine. 2015;21(9):989–97. [DOI] [PubMed] [Google Scholar]
- 41.Nlandu-Khodo S, Neelisetty S, Phillips M, et al. Blocking TGF-beta and beta-Catenin Epithelial Crosstalk Exacerbates CKD. J Am Soc Nephrol 2017;28(12):3490–503.**Deletion of TbRII in proximal tubule epithelial cells paradoxically increased tubular apoptosis and fibrosis in models of chronic kidney disease. These cells had reduced b-catenin activity and was rescued by conditional stabilization of b-catenin in vivo. Augmenting b-catenin signaling in aristolochic acid-treated proximal tubule cells lacking TbRII in vitro reduced both apoptosis and G2/M arrest.
- 42.Sureshbabu A, Muhsin SA, Choi ME. TGF-beta signaling in the kidney: profibrotic and protective effects. Am J Physiol Renal Physiol 2016;310(7):F596–F606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Yang L, Besschetnova TY, Brooks CR, et al. Epithelial cell cycle arrest in G2/M mediates kidney fibrosis after injury. Nature medicine. 2010;16(5):535–43, 1p following 143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Alvarez RJ, Sun MJ, Haverty TP, et al. Biosynthetic and proliferative characteristics of tubulointerstitial fibroblasts probed with paracrine cytokines. Kidney Int 1992;41(1):14–23. [DOI] [PubMed] [Google Scholar]
- 45.Ma LJ, Jha S, Ling H, et al. Divergent effects of low versus high dose anti-TGF-beta antibody in puromycin aminonucleoside nephropathy in rats. Kidney Int 2004;65(1):106–15. [DOI] [PubMed] [Google Scholar]
- 46.Urakawa I, Yamazaki Y, Shimada T, et al. Klotho converts canonical FGF receptor into a specific receptor for FGF23. Nature. 2006;444(7120):770–4. [DOI] [PubMed] [Google Scholar]
- 47.Kurosu H, Yamamoto M, Clark JD, et al. Suppression of aging in mice by the hormone Klotho. Science. 2005;309(5742):1829–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Koh N, Fujimori T, Nishiguchi S, et al. Severely reduced production of klotho in human chronic renal failure kidney. Biochemical and biophysical research communications. 2001;280(4):1015–20. [DOI] [PubMed] [Google Scholar]
- 49.Ohyama Y, Kurabayashi M, Masuda H, et al. Molecular cloning of rat klotho cDNA: markedly decreased expression of klotho by acute inflammatory stress. Biochemical and biophysical research communications. 1998;251(3):920–5. [DOI] [PubMed] [Google Scholar]
- 50.Irifuku T, Doi S, Sasaki K, et al. Inhibition of H3K9 histone methyltransferase G9a attenuates renal fibrosis and retains klotho expression. Kidney Int 2016;89(1):147–57. [DOI] [PubMed] [Google Scholar]
- 51.Yin S, Zhang Q, Yang J, et al. TGFbeta-incurred epigenetic aberrations of miRNA and DNA methyltransferase suppress Klotho and potentiate renal fibrosis. Biochim Biophys Acta Mol Cell Res 2017;1864(7):1207–16. [DOI] [PubMed] [Google Scholar]
- 52.Humphreys BD, Lin SL, Kobayashi A, et al. Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol 2010;176(1):85–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kramann R, Dirocco DP, Maarouf OH, et al. Matrix Producing Cells in Chronic Kidney Disease: Origin, Regulation, and Activation. Curr Pathobiol Rep 2013;1(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Sasaki K, Doi S, Nakashima A, et al. Inhibition of SET Domain-Containing Lysine Methyltransferase 7/9 Ameliorates Renal Fibrosis. J Am Soc Nephrol 2016;27(1):203–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Tamura R, Doi S, Nakashima A, et al. Inhibition of the H3K4 methyltransferase SET7/9 ameliorates peritoneal fibrosis. PloS one. 2018;13(5):e0196844.*An inhibitor of the H3K4 methyltransferase SET7/9, sinefungin, reduced fibrosis in a murine model of peritoneal fibrosis. Sinefungin also reduced TGF-b1-stimulated fibrosis and H3K4 mono-methylation. SET7/9 expression was upregulated in nonadherent cells from effluent of peritoneal dialysis patients compared with other human peritoneal mesothelial cells.
- 56.Smith ER, Holt SG, Hewitson TD. FGF23 activates injury-primed renal fibroblasts via FGFR4-dependent signalling and enhancement of TGF-beta autoinduction. Int J Biochem Cell Biol 2017;92:63–78. [DOI] [PubMed] [Google Scholar]
- 57.Smith ER, Tan SJ, Holt SG, et al. FGF23 is synthesised locally by renal tubules and activates injury-primed fibroblasts. Sci Rep 2017;7(1):3345.*Fibroblasts isolated from UUO kidneys, but not NRKF (normal rat kidney fibroblasts), responded to FGF23 with increases in Col1a1 and protein levels of a-SMA. These responses were partially suppressed by inhbiting TGF-b using siRNA.
- 58.Szeto SG, Narimatsu M, Lu M, et al. YAP/TAZ Are Mechanoregulators of TGF-beta-Smad Signaling and Renal Fibrogenesis. J Am Soc Nephrol 2016;27(10):3117–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Miranda MZ, Bialik JF, Speight P, et al. TGF-beta1 regulates the expression and transcriptional activity of TAZ protein via a Smad3-independent, myocardin-related transcription factor-mediated mechanism. J Biol Chem 2017;292(36):14902–20.*TGF-b signaling increased the transcription, but not localization, of TAZ in both fibroblast and epithelial cell lines. The mechanism was through a p38-mediated increasae in myocardin-related transcription factor (MRTF). This effect primed cells for augmented TAZ activity upon a second, activating stimulus.
- 60.Anorga S, Overstreet JM, Falke LL, et al. Deregulation of Hippo-TAZ pathway during renal injury confers a fibrotic maladaptive phenotype. FASEB J 2018;32(5):2644–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Basile DP, Donohoe D, Roethe K, et al. Renal ischemic injury results in permanent damage to peritubular capillaries and influences long-term function. Am J Physiol Renal Physiol 2001;281(5):F887–99. [DOI] [PubMed] [Google Scholar]
- 62.Nakagawa T, Li JH, Garcia G, et al. TGF-beta induces proangiogenic and antiangiogenic factors via parallel but distinct Smad pathways. Kidney Int 2004;66(2):605–13. [DOI] [PubMed] [Google Scholar]
- 63.Xavier S, Vasko R, Matsumoto K, et al. Curtailing endothelial TGF-beta signaling is sufficient to reduce endothelial-mesenchymal transition and fibrosis in CKD. J Am Soc Nephrol 2015;26(4):817–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Kinashi H, Ito Y, Sun T, et al. Roles of the TGF-beta(-)VEGF-C Pathway in Fibrosis-Related Lymphangiogenesis. Int J Mol Sci 2018;19(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Sakamoto I, Ito Y, Mizuno M, et al. Lymphatic vessels develop during tubulointerstitial fibrosis. Kidney Int 2009;75(8):828–38. [DOI] [PubMed] [Google Scholar]
- 66.Jeltsch M, Kaipainen A, Joukov V, et al. Hyperplasia of lymphatic vessels in VEGF-C transgenic mice. Science. 1997;276(5317):1423–5. [DOI] [PubMed] [Google Scholar]
- 67.Oka M, Iwata C, Suzuki HI, et al. Inhibition of endogenous TGF-beta signaling enhances lymphangiogenesis. Blood. 2008;111(9):4571–9. [DOI] [PubMed] [Google Scholar]
- 68.Clavin NW, Avraham T, Fernandez J, et al. TGF-beta1 is a negative regulator of lymphatic regeneration during wound repair. Am J Physiol Heart Circ Physiol 2008;295(5):H2113–27. [DOI] [PubMed] [Google Scholar]
- 69.Suzuki Y, Ito Y, Mizuno M, et al. Transforming growth factor-beta induces vascular endothelial growth factor-C expression leading to lymphangiogenesis in rat unilateral ureteral obstruction. Kidney Int 2012;81(9):865–79. [DOI] [PubMed] [Google Scholar]
- 70.Kinashi H, Falke LL, Nguyen TQ, et al. Connective tissue growth factor regulates fibrosis-associated renal lymphangiogenesis. Kidney Int 2017;92(4):850–63.**Increased lymphangiogenesis was associated with increased CTGF and VEGF-C in human chronic kidney disease. In CTGF knockout mice, lymphangiogenesis was reduced after two models of injury, UUO and ischemia/reperfusion. In vitro, TGF-b-induced VEGF-C production was abrogated with CTGF knockdown. A direct interaction between CTGF and VEGF-C was shown with surface plasmon resonance analysis.

