Elsevier

Life Sciences

Volume 90, Issues 13–14, 9 April 2012, Pages 509-518
Life Sciences

High glucose enhances TGF-β1 expression in rat bone marrow stem cells via ERK1/2-mediated inhibition of STAT3 signaling

https://doi.org/10.1016/j.lfs.2012.01.005Get rights and content

Abstract

Aims

This study was to investigate the effect of high glucose (HG) on TGF-β1 expression and the underlying mechanisms in bone marrow stem cells.

Main methods

Rat bone marrow multipotent adult progenitor cells (MAPCs) were cultured in normal (5.5 mM d-glucose) and HG media (25.5 mM d-glucose) for up to 14 days. l-Glucose (20 mM plus 5.5 mM d-glucose) was used as high osmolarity control. TGF-β1 expression was evaluated using quantitative RT-PCR, ELISA, and immunofluorescence staining for its mRNA and protein level in the cells and in the conditioned media. The expression and activation of ERK1/2 and STAT3 were examined in MAPCs cultured in HG media with Western blot.

Key findings

Measurable level of TGF-β1 was detected in the cells cultured in normal media. TGF-β1 expression was substantially increased in MAPCs after 36 h of culture in HG media with over 20-fold increase in the mRNA and 5-fold increase in protein level over control. Interestingly, ERK1/2 phosphorylation was significantly increased in MAPCs cultured in HG media, while in STAT3 (Tyr705), not STAT3 (Ser727), phosphorylation was dramatically decreased. Treatment of cells with the specific MEK1 inhibitor PD98059 or U0126 suppressed ERK1/2 phosphorylation and TGF-β1 expression, and completely restored the level of STAT3 (Tyr705) phosphorylation in MAPCs cultured in HG media. Treatment of the cells with the specific STAT3 phosphorylation inhibitor AG490 significantly blocked STAT3 (Tyr705) phosphorylation and increased TGF-β1 expression without change in ERK1/2 phosphorylation in MPACs.

Significance

HG increased TGF-β1 expression through inhibition of STAT3 (Tyr705) by enhanced ERK1/2 signaling in MAPCs.

Introduction

Cell therapy with bone marrow mesenchymal stem cells (MSCs) remains a viable option for tissue repair and regeneration after injuries including myocardial infarction (Williams and Hare, 2011). While the mechanisms for the beneficial effects of MSCs on cardiac repair are complex and not fully understood, paracrine signaling is believed to be responsible (at least partially) for their beneficial effects including myogenesis, angiogenesis, and collateral development (Mirotsou et al., 2011). A wide spectrum of biologically active growth factors and cytokines including, but not limited to, fibroblast growth factor (FGF), interleukin-1 and 6, transforming growth factor-β (TGF-β), and VEGF are generated from MSCs and released into the culture medium (Mirotsou et al., 2011). It has been shown that cardiovascular risk factors such as hypercholesterolemia and diabetes mellitus (DM) attenuate native collateral development and are associated with incomplete revascularization and decreased re-endothelialization after arterial injury in both human and animals (Ii et al., 2006, Kinnaird et al., 2008, Sodha et al., 2009). The mechanisms for the impaired outcome in the patients with hypercholesterolemia and/or DM have not been well defined. It is possible that these risk factors like DM may impair the function of MSCs involved in cardiovascular repair and regeneration. Indeed, bone marrow-derived progenitor cells from patients with hypercholesterolemia and/or DM exhibit a substantially reduced capacity for neovascularization and decreased paracrine secretion of proangiogenic factors (Tepper et al., 2002, Loomans et al., 2004, Kränkel et al., 2005, Heeschen et al., 2004). Recently, we demonstrated that high glucose (HG) substantially suppressed VEGF expression in MAPCs through inhibition of JAK2/STAT3 signaling (Liu et al., 2008).

TGF-β is the prototypic member of a super family of multifunctional cytokines involved in many cellular functions including cell growth, migration, adhesion, differentiation, and angiogenesis (Iruela-Arispe and Sage, 1993, Schmierer and Hill, 2007). The super family has thirty-three members in mammals including three TGF-β isoforms (TGF-β1, TGF-β2, and TGF-β3), activins, and bone morphogenetic proteins (BMPs) (Schmierer and Hill, 2007). Perturbations in TGF-β signaling are associated with cardiovascular diseases such as impaired vascular development and angiogenesis (Grainger, 2007, Goumans et al., 2009). TGF-β1 is produced in various cells including endothelial cells and MSCs (Vinals and Pouyssegur, 2001, Worster et al., 2000). Multipotent adult progenitor cells (MAPCs) are clonally isolated from postnatal human and rodent tissues including bone marrow, muscle and brain (Jiang et al., 2002a, Jiang et al., 2002b). These cells have been well characterized and are able to differentiate into multiple cell lineages including endothelial cells, hepatocytes, cardiomyocytes, and neurons (Jiang et al., 2002a, Jiang et al., 2002b, Ulloa-Montoya et al., 2007, Lu et al., 2010). MAPCs promote angiogenesis and improve cardiac and limb function when injected into the peri-infarct areas in ischemic mice (Pelacho et al., 2007, Aranguren et al., 2008). Interestingly, the increased vascularity and improved cardiac function after myocardial infarction is not due to differentiation and direct contribution of MAPCs to the vascular or cardiomyocyte compartment after cell transplantation. Instead, the beneficial effects are largely contributed by secreting vascular growth factors like VEGF and TGF-β by these cells, resulting in increased angiogenesis and cardioprotection (Pelacho et al., 2007, Agbulut et al., 2006). MSCs have also been shown to enhance wound healing at least partially due to the release of different cytokines and chemokines, such as TGF-β1 and VEGF (Chen et al., 2008). Therefore, optimal expression of these growth factors and cytokines is critical to the optimal outcome of cell therapies especially in the patients with DM that is associated with significant cardiovascular morbidity and mortality (Engelgau et al., 2004).

In the present study, we investigated the effect of HG on the expression of TGF-β1 and the underlying mechanism in rat MAPCs. We observed that detectable level of TGF-β1 was expressed in rat MAPCs with balanced phosphorylation of both ERK1/2 and STAT3. TGF-β1 expression was substantially increased in MAPCs cultured in HG media. HG also significantly enhanced ERK1/2 phosphorylation, and inhibited STAT3 (Tyr705) phosphorylation in MAPCs. Treatment of cells with the specific MEK1 inhibitor PD98059 or U0126 remarkably suppressed ERK1/2 phosphorylation and TGF-β1 expression, and completely restored the level of STAT3 (Tyr705) phosphorylation in MAPCs cultured in HG media. Treatment of the cells with the specific STAT3 phosphorylation inhibitor AG490 significantly blocked STAT3 (Tyr705) phosphorylation, and increased TGF-β1 expression without change in ERK1/2 phosphorylation. These data suggested that HG increased TGF-β1 expression through inhibition of STAT3 (Tyr705) by enhanced ERK1/2 signaling in MAPCs.

Section snippets

Materials

Low-Glucose Dulbecco's minimal essential medium (DMEM) was obtained from Gibco BRL (Grand Island, NY, USA). Fibronectin (FN), MCDB-201, insulin–transferrin–selenium (ITS), linoleic-acid-bovine-serum-albumin (LA-BSA), dexamethasone, l-ascorbic acid 2-phosphate sesquimagnesium salt (l-ascorbic acid), and epidermal growth factor (EGF) were purchase from Sigma (St. Louis, MO). Platelet-derived growth factor-BB (PDGF-BB) and anti-VEGF mouse antibody were from R&D Systems (Minneapolis, MN, USA).

High glucose dramatically enhanced TGF-β1 expression in MAPCs

We first evaluated the transcriptional expression of TGF-β1 gene as reflected by mRNA level in MAPCs using RT-PCR under normal culture condition. There was a low, but detectable level of transcriptional expression of TGF-β1 in MAPCs in normal media (5.5 mM d-glucose) (Fig. 1A). No changes in TGF-β1 expression was observed in the cells during culture for up to 96 h. However, the TGF-β1 mRNA level was significantly increased in MAPCs exposed to HG (25.5 mM d-glucose) by almost 3 times over control

Discussion

TGF-β1 is expressed in many types of cells including endothelial cells, smooth muscle cells, mesangial cell and MSCs (Vinals and Pouyssegur, 2001, Redondo et al., 2007, Kim et al., 2007a, Kim et al., 2007b, Worster et al., 2000). The regulation of TGF-β1 expression and function is very complex and tightly controlled, and involves a variety of factors including angiotensin II (Ang II), aldosterone (Han et al., 2009), basic fibroblast growth factor (Dhandapani et al., 2007), tumor necrosis factor

Conflict of interest statement

No conflict of interest in any form.

Acknowledgments

This work was supported by a grant from the Natural Science Foundation of China for the Youth (Number 81000337 for ZHL), and NIH R01 HL094650 (ZGL).

References (59)

  • O. Agbulut et al.

    Can bone marrow-derived multipotent adult progenitor cells regenerate infarcted myocardium?

    Cardiovasc Res

    (2006)
  • X.L. Aranguren et al.

    Multipotent adult progenitor cells sustain function of ischemic limbs in mice

    J Clin Invest

    (2008)
  • S. Belmadani et al.

    Microvessel vascular smooth muscle cells contribute to collagen type I deposition through ERK1/2 MAP kinase, alphavbeta3-integrin, and TGF-beta1 in response to ANG II and high glucose

    Am J Physiol Heart Circ Physiol

    (2008)
  • L. Chen et al.

    Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing

    PLoS One

    (2008)
  • J. Chung et al.

    STAT3 serine phosphorylation by ERK-dependent and -independent pathways negatively modulates its tyrosine phosphorylation

    Mol Cell Biol

    (1997)
  • B.D. Cook et al.

    TGF-beta1 induces rearrangement of FLK-1-VE-cadherin-beta-catenin complex at the adherens junction through VEGF-mediated signaling

    J Cell Biochem

    (2008)
  • K.M. Dhandapani et al.

    Induction of transforming growth factor-beta1 by basic fibroblast growth factor in rat C6 glioma cells and astrocytes is mediated by MEK/ERK signaling and AP-1 activation

    J Neurosci Res

    (2007)
  • M.M. Engelgau et al.

    The evolving diabetes burden in the United States

    Ann Intern Med

    (2004)
  • G. Ferrari et al.

    VEGF, a prosurvival factor, acts in concert with TGF-beta1 to induce endothelial cell apoptosis

    Proc Natl Acad Sci U S A

    (2006)
  • G. Ferrari et al.

    Transforming growth factor-beta 1 (TGF-beta1) induces angiogenesis through vascular endothelial growth factor (VEGF)-mediated apoptosis

    J Cell Physiol

    (2009)
  • M.A. Frias et al.

    Native and reconstituted HDL activate Stat3 in ventricular cardiomyocytes via ERK1/2: role of sphingosine-1-phosphate

    Cardiovasc Res

    (2009)
  • H. Fujita et al.

    ERK and p38 mediate high-glucose-induced hypertrophy and TGF-beta expression in renal tubular cells

    Am J Physiol Renal Physiol

    (2004)
  • M. Giannopoulou et al.

    Distinctive role of Stat3 and Erk-1/2 activation in mediating interferon-gamma inhibition of TGF-beta1 action

    Am J Physiol Renal Physiol

    (2006)
  • M.J. Goumans et al.

    TGF-beta signaling in vascular biology and dysfunction

    Cell Res

    (2009)
  • D.J. Grainger

    TGF-beta and atherosclerosis in man

    Cardiovasc Res

    (2007)
  • D. Gramatzki et al.

    Aryl hydrocarbon receptor inhibition downregulates the TGF-beta/Smad pathway in human glioblastoma cells

    Oncogene

    (2009)
  • G.H. Guo et al.

    Hepatitis B virus X protein promotes proliferation and upregulates TGF-beta1 and CTGF in human hepatic stellate cell line, LX-2

    Hepatobiliary Pancreat Dis Int

    (2009)
  • J.S. Han et al.

    Aldosterone-induced TGF-beta1 expression is regulated by mitogen-activated protein kinases and activator protein-1 in mesangial cells

    J Korean Med Sci

    (2009)
  • T. Hayashida et al.

    High ambient glucose enhances sensitivity to TGF-beta1 via extracellular signal-regulated kinase and protein kinase C delta activities in human mesangial cells

    J Am Soc Nephrol

    (2004)
  • Cited by (6)

    View full text