Elsevier

Differentiation

Volume 92, Issue 3, September 2016, Pages 102-107
Differentiation

Review article
The origin of renal fibroblasts/myofibroblasts and the signals that trigger fibrosis

https://doi.org/10.1016/j.diff.2016.05.008Get rights and content

Abstract

Renal fibrosis is a common characteristic of chronic kidney disease (CKD). Aberrant and excessive depositions of extracellular matrix (ECM) proteins in both glomeruli and interstitial regions are typical hallmarks of renal fibrosis and amplify the severity of kidney injury. To date, an approved therapy specifically targeted to renal fibrosis is needed to mitigate or even retard renal fibrosis. Recent findings have identified a unique population of myofibroblasts as a primary source of ECM in scar tissue formation. However, the origin of myofibroblasts in renal fibrosis remains the subject of controversial debates. The advancement in lineage tracing and immunofluorescent microscopy technologies have suggested that myofibroblasts may arise from a number of sources such as activated renal fibroblasts, pericytes, epithelial-to-mesenchymal transition (EMT), endothelial-to-mesenchymal transition (EndoMT), bone marrow derived cells and fibrocytes. Recent studies also indicate that multiple ligands of TGF-β/Smads are the direct mediators for renal fibrosis. Consistently, inhibition of the TGF-β/Smads signaling pathway using various strategies significantly reduce renal fibrotic lesions and ameliorate kidney injury, suggesting that targeting the TGF-β/Smads signaling pathway could be a new strategy for effective therapies. In this review, we will briefly discuss the diverse origins of myofibroblasts and molecular pathways triggering renal fibrosis. Prospective therapeutic approaches based on those molecular mechanisms will hopefully offer exciting insights in the development of new therapeutic interventions for patients in the near future.

Introduction

The epidemic of chronic kidney disease (CKD) has been rising significantly over the past few decades and become a major health problem imposing enormous socioeconomic challenges to society. The causes of CKD can be secondary to diseases such as diabetes, hypertension, HIV infection and obesity, or due to primary kidney injury such as focal segmental glomerulosclerosis (FSGS) and acute kidney injury (AKI) (Mikulak and Singhal, 2010, Quigley et al., 2009, Venkatachalam et al., 2015, Nakagawa et al., 2007, Gasser et al., 2013, Sarafidis, 2008). All these pathological events cause normal kidney parenchymal destruction with progressive scar tissue formation, eventually leading to renal fibrosis, which is a common characteristic of end-stage renal disease. Fibrosis is marked by excessive and pathological accumulation of extracellular matrix (ECM) consisting of collagen I, III and IV, fibronectin, laminin, heparan and perlecan. Such aberrant ECM accumulation can result in destruction of an organ accompanied with compromised organ function. glomerulosclerosis and tubulointerstitial fibrosis are the common hallmarks of CKD. Fibrotic lesions in the glomerular tuft consist mainly of collagen I and IV and fibronectin, resulting in loss of blood flow and reduced filtration. ECM accumulation in the region between tubules and peritubular capillaries impede tubular normal function involving the removal of toxins and mediating cellular transport. As the fibrosis expands to the surrounding capillaries and the entire nephron, the kidney loses its structural support and a decrease in kidney volume results in loss of renal function. Currently, the common therapeutic strategy for treating non-diabetic CKD is the control of systemic blood pressure by blocking the angiotensin-aldosterone system (RAAS) to reduce proteinuria and slow the progression of CKD (Thethi et al., 2012, Choudhury et al., 2010). To date, an approved therapy specifically targeted to renal fibrosis is not available, even though extensive research has been performed to investigate the primary molecular mediators of fibrosis. Research involving animal models of kidney disease has identified myofibroblasts as major matrix producing cells contributing to fibrosis after injury.

Myofibroblasts can be characterized by being α-smooth muscle actin (α-SMA) positive, are mainly located in the renal interstitium and to a lesser extent in glomeruli in various animal models of renal fibrosis. The percentage of myofibroblasts is proportionally correlated with severity of renal fibrosis. However, the diverse origin and mixed phenotypic heterogeneity of myofibroblasts makes them a difficult therapeutic target. Therefore, delineating the origin of myofibroblasts and their molecular signaling pathways would provide valuable information for designing effective therapies to stop or even reverse renal fibrosis.

Section snippets

Fibroblasts

Fibroblasts are quiescent cells found within the interstitial space, important for maintaining the structural integrity of kidneys by producing basal level of extracellular matrix. During the normal tissue repair process after damage, fibroblasts are activated by inflammatory cytokines, growth factors, hypoxia and mechanical forces to proliferate and produce collagens to make new connective tissue. However, in the process of fibrosis, it is believed that aberrant fibroblast activation and the

Pericytes

The recent discovery of a functionally distinct subset of extracellular matrix producing cells called pericytes, might be another major source of myofibroblasts. Pericytes are stromal cells in close contact with interstitial capillary endothelial cells to regulate capillary permeability. The identification of pericytes and their contribution to fibrogenesis in kidney fibrosis are highly controversial due to lack of specific cellular markers and of strategies in lineage tracing. Lin et al. (2008)

EMT

The essential role of Epithelial-Mesenchymal Transition (EMT) during developmental processes, wound healing, organ fibrosis and cancer metastasis has been extensively studied in past decades (Yang et al., 2010, Liu et al., 2014, Lovisa et al., 2015, Iwano et al., 2002, Inoue et al., 2015, Li et al., 2008, Velden et al., 2011). It is generally accepted that epithelial cells lose their cell-cell adhesion and cell polarity properties by diminishing important adhesion markers like E-cadherin,

EndoMT

Endothelial-Mesenchymal Transition (EndoMT) is a unique subset of EMT, a process similar to EMT, whereby endothelial cells gradually lose their markers and acquire mesenchymal markers. Lineage tracing of endothelial cells in various organs from different disease models has demonstrated an important mechanism in the pathogenesis of fibrosis. The essential role and contribution of endothelial cells to the myofibroblast pool in kidney fibrosis is largely unknown and debatable. Zeisberg et al.

Bone marrow-derived myofibroblasts

Myofibroblasts may originate from bone marrow-derived cells (BMDC) migrating into the kidney in response to injury. Sex-mismatched kidney transplantation in humans demonstrated circulating mesenchymal precursor cells have the potential to migrate to areas of inflammation (Yamashita et al., 2012). In the UUO model of kidney fibrosis, transplantation of transgenic bone marrow expressing red fluorescent protein (RFP) under the control of α-SMA promoter (α-SMA-RFP) demonstrated 35% of the

Fibrocytes

Hematopoietic or bone marrow-derived collagen producing fibrocytes expressing vimentin and CD34, infiltrate renal parenchymal and contribute to fibrogenesis. Patients with chronic allograft nephropathy with interstitial fibrosis had a large number of myofibroblasts derived from the recipient. Immunostaining of hematopoietic markers like CD11b, CD34, CD45, CD115, Gr1, together with intracellular collagen I were used to detect fibrocytes in kidneys (Cvetkovic et al., 2005, Acevedo et al., 2008,

Signals triggering fibrosis

The exact molecular mechanisms of fibrogenesis in the kidney remain largely unknown. Understanding and delineation of signaling pathways that trigger fibrosis would aid in designing new therapeutic approaches to prevent or even reverse renal fibrosis. Multiple signaling pathways are involved in renal fibrosis, such as TGF-β/Smads, Wnt/β-catenin, JNK/STAT3, and MAPKs (Yu et al., 2007, Sun et al., 2013, Maarouf et al., 2015, Furukawa et al., 2003, Lim et al., 2009). Increasing evidence has

New targets in anti-fibrotic treatment of CKD

Kidney fibrosis is a pathological characteristic of the end-point of chronic kidney disease independent of aetiology. None of the current therapies is effective to reduce kidney fibrosis and to stop the disease progression. With current advances in research of molecular pathways and targets, multiple therapeutic strategies have been proposed to prevent or even degrade fibrotic tissues in kidneys. Fibrosis is often associated with robust inflammatory reactions and immune cell infiltration.

Conclusion

In summary, numerous works published over the past decades established that kidney fibrosis is a complex and dynamic process involving all different types of cells within the kidney (Fig. 1). With the current understanding of kidney fibrosis progression, it is hypothesized that inhibition or removal of myofibroblasts will decrease kidney fibrosis and improve renal function. However, targeting the diverse origins and phenotypic heterogeneity of myofibroblasts is the major challenge in designing

Conflict of interest statement

The authors have no conflict of interest to declare.

Acknowledgments

This study was supported by the National Health and Medical Research Council (NHMRC) of Australia (ID606540 and APP1057581).

References (70)

  • J.H. Li et al.

    Advanced glycation end products induce tubular epithelial-myofibroblast transition through the RAGE-ERK1/2 MAP kinase signaling pathway

    Am. J. Pathol.

    (2004)
  • Y. Li et al.

    Epithelial-to-mesenchymal transition is a potential pathway leading to podocyte dysfunction and proteinuria

    Am. J. Pathol.

    (2008)
  • S.L. Lin et al.

    Pericytes and perivascular fibroblasts are the primary source of collagen-producing cells in obstructive fibrosis of the kidney

    Am. J. Pathol.

    (2008)
  • X. Liu et al.

    Epidermal growth factor inhibits transforming growth factor-β-induced fibrogenic differentiation marker expression through ERK activation

    Cell. Signal.

    (2014)
  • Y. Liu

    Cellular and molecular mechanisms of renal fibrosis

    Nat. Rev. Nephrol.

    (2011)
  • M. Mariasegaram et al.

    Lefty antagonises TGF-beta1 induced epithelial-mesenchymal transition in tubular epithelial cells

    Biochem. Biophys. Res. Commun.

    (2010)
  • X.-M. Meng et al.

    Disruption of Smad4 impairs TGF-b/Smad3 and Smad7 transcriptional regulation during renal inflammation and fibrosis in vivo and in vitro

    Kidney Int.

    (2012)
  • S. Neelisetty et al.

    Renal fibrosis is not reduced by blocking transforming growth factor-β signaling in matrix-producing interstitial cells

    Kidney Int.

    (2015)
  • D.J. Nikolic-Paterson et al.

    Macrophages promote renal fibrosis through direct and indirect mechanisms

    Kidney Int. Suppl.

    (2014)
  • X. Qu et al.

    Regulation of renal fibrosis by Smad3 Thr388 phosphorylation

    Am. J. Pathol.

    (2014)
  • X. Qu et al.

    The Smad3/Smad4/CDK9 complex promotes renal fibrosis in mice with unilateral ureteral obstruction

    Kidney Int.

    (2015)
  • R. Samarakoon et al.

    Induction of renal fibrotic genes by TGF-β1 requires EGFR activation, p53 and reactive oxygen species

    Cell. Signal.

    (2013)
  • K. Tamaki et al.

    TGF-beta 1 in glomerulosclerosis and interstitial fibrosis of adriamycin nephropathy

    Kidney Int.

    (1994)
  • R.J. Tan et al.

    Wnt/β-catenin signaling and kidney fibrosis

    Kidney Int. Suppl.

    (2014)
  • M.A. Venkatachalam et al.

    Fibrosis without fibroblast TGF-β receptors?

    Kidney Int.

    (2015)
  • C.F. Wu et al.

    Transforming growth factor β-1 stimulates profibrotic epithelial signaling to activate pericyte-myofibroblast transition in obstructive kidney fibrosis

    Am. J. Pathol.

    (2013)
  • L.M. Acevedo et al.

    Glomerular CD34 expression in short- and long-term diabetes

    J. Histochem. Cytochem.

    (2008)
  • D. Choudhury et al.

    Diabetic nephropathy – a multifaceted target of new therapies

    Discov. Med.

    (2010)
  • A.C.K. Chung et al.

    Advanced glycation end-products induce tubular CTGF via TGF-beta-independent Smad3 signaling

    J. Am. Soc. Nephrol.

    (2010)
  • A.C.K. Chung et al.

    Disruption of the Smad7 gene promotes renal fibrosis and inflammation in unilateral ureteral obstruction (UUO) in mice

    Nephrol. Dial. Transpl.

    (2009)
  • I. Cvetkovic et al.

    Critical role of macrophage migration inhibitory factor activity in experimental autoimmune diabetes

    Endocrinology

    (2005)
  • D.L. Gasser et al.

    Focal segmental glomerulosclerosis is associated with a PDSS2 haplotype and independently, with a decreased content of coenzyme Q10

    AJP: Ren. Physiol.

    (2013)
  • M.T. Grande et al.

    Snail1-induced partial epithelial-to-mesenchymal transition drives renal fibrosis in mice and can be targeted to reverse established disease

    Nat. Med.

    (2015)
  • J. Guo et al.

    RAGE mediates podocyte injury in adriamycin-induced glomerulosclerosis

    J. Am. Soc. Nephrol.

    (2008)
  • X.R. Huang et al.

    Mice overexpressing latent TGF-beta1 are protected against renal fibrosis in obstructive kidney disease

    AJP: Ren. Physiol.

    (2008)
  • Cited by (279)

    View all citing articles on Scopus
    View full text