Transforming growth factor beta (TGF-β) and inflammation in cancer

https://doi.org/10.1016/j.cytogfr.2009.11.008Get rights and content

Abstract

The transforming growth factor beta (TGF-β) has been studied with regard to the regulation of cell behavior for over three decades. A large body of research has been devoted to the regulation of epithelial cell and derivative carcinoma cell populations in vitro and in vivo. TGF-β has been shown to inhibit epithelial cell cycle progression and promote apoptosis that together significantly contribute to the tumor suppressive role for TGF-β during carcinoma initiation and progression. TGF-β is also able to promote an epithelial to mesenchymal transition that has been associated with increased tumor cell motility, invasion and metastasis. However, it has now been shown that loss of carcinoma cell responsiveness to TGF-β stimulation can also promote metastasis. Interestingly, enhanced metastasis in the absence of a carcinoma cell response to TGF-β stimulation has been shown to involve increased chemokine production resulting in recruitment of pro-metastatic myeloid derived suppressor cell (MDSC) populations to the tumor microenvironment at the leading invasive edge. When present, MDSCs enhance angiogenesis, promote immune tolerance and provide matrix degrading enzymes that promote tumor progression and metastasis. Further, the recruitment of MDSC populations in this context likely enhances the classic role for TGF-β in immune suppression since the MDSCs are an abundant source of TGF-β production. Importantly, it is now clear that carcinoma-immune cell cross-talk initiated by TGF-β signaling within the carcinoma cell is a significant determinant worth consideration when designing therapeutic strategies to manage tumor progression and metastasis.

Section snippets

TGF-β dependent regulation of epithelial cell populations

TGF-β was initially described as a ligand that had the ability to promote or suppress cellular proliferation depending on the cell type and context of stimulation. In non-transformed or carcinoma-associated epithelial cells, TGF-β is well known for its ability to inhibit cell proliferation and promote an epithelial to mesenchymal transition (EMT) that has been associated with increased motility and invasiveness [56], [57]. In the years since discovery of TGF-β, an enormous volume of data has

Immature bone marrow derived cells promote invasion and metastasis

In many of the early studies, it was suggested that the loss of TGF-β signaling in carcinoma cells could lead to enhanced growth of primary tumors and metastases. However, it had also been suggested that TGF-β signaling was necessary for promoting an epithelial to mesenchymal transition that was thought to be essential for metastatic dissemination. This latter point was difficult to reconcile with data demonstrating that increased TGF-β signaling or complete abrogation of TGF-β signaling in

T-cells are significantly regulated by TGF-β

The impact of TGF-β signaling in the immune system is well documented. Importantly, TGF-β is present in the carcinoma extra-cellular matrix regardless of the carcinoma cell specific ability to bind the activated ligand. Further, in tumors where the carcinoma cells were unable to respond to TGF-β, recent results suggest that stromal TGF-β was more abundant [76]. Thus, in carcinomas where the carcinoma cell specific response to TGF-β is attenuated or ablated there remains an abundant supply of

TGF-β attenuates NK-cell associated tumor rejection

TGF-β inhibits NK cell and neutrophil effector functions and thereby contributes to a permissive microenvironment for tumor progression (Fig. 5B). In early studies, it was shown that TGF-β stimulation resulted in suppression of natural killer (NK) cell activity. in vivo, it was shown that growth of MDA-MB-231 mammary carcinomas and derivative metastases could be significantly reduced when systemic TGF-β inhibition was performed [107]. This effect was not observed in beige nude mice that were NK

TGF-β regulates monocyte and macrophage activity

Monocytes and macrophages have been studied in great detail with regard to their contribution to tumor progression. In general, the recruitment of monocytes and macrophages to the tumor microenvironment has been associated with enhanced tumor progression. TGF-β promotes recruitment of monocytes and it has been suggested that TGF-β can promote monocyte to macrophage differentiation [119], [120] (Fig. 5B). Early work suggested that in the case of macrophages that have the potential to be

Summary

The loss of TGF-β signaling in many common human carcinomas has been well documented [16]. Abundant literature dedicated to the impact of TGF-β signaling in non-transformed and carcinoma associated epithelium has focused primarily on the regulation of carcinoma cell behavior. Clearly the regulation of carcinoma cell survival, proliferation and epithelial to mesenchymal transition are significant factors that contribute to TGF-β mediated regulation of tumor progression. The current literature

Brian Bierie, Ph.D. performed his undergraduate work at Colorado State University in his home town of Fort Collins, Colorado. After finishing his undergraduate work he spent time working in the area of analytical chemistry for a large pesticide production plant. During this time he realized that some of the mutagens were so abundant in the soil and waste water produced by the production facilities that he began to think about dedicating his time to studies that could help people rather than

References (129)

  • H. Hayashi et al.

    The MAD-related protein Smad7 associates with the TGFbeta receptor and functions as an antagonist of TGFbeta signaling

    Cell

    (1997)
  • P. Kavsak et al.

    Smad7 binds to Smurf2 to form an E3 ubiquitin ligase that targets the TGF beta receptor for degradation

    Mol Cell

    (2000)
  • T. Ebisawa et al.

    Smurf1 interacts with transforming growth factor-beta type I receptor through Smad7 and induces receptor degradation

    J Biol Chem

    (2001)
  • C.H. Heldin et al.

    Mechanism of TGF-beta signaling to growth arrest, apoptosis, and epithelial-mesenchymal transition

    Curr Opin Cell Biol

    (2009)
  • C.W. Daniel et al.

    TGF-beta 1-induced inhibition of mouse mammary ductal growth: developmental specificity and characterization

    Dev Biol

    (1989)
  • E.C. Kordon et al.

    Ectopic TGF beta 1 expression in the secretory mammary epithelium induces early senescence of the epithelial stem cell population

    Dev Biol

    (1995)
  • K.B. Ewan et al.

    Latent transforming growth factor-beta activation in mammary gland: regulation by ovarian hormones affects ductal and alveolar proliferation

    Am J Pathol

    (2002)
  • A.E. Gorska et al.

    Transgenic mice expressing a dominant-negative mutant type II transforming growth factor-beta receptor exhibit impaired mammary development and enhanced mammary tumor formation

    Am J Pathol

    (2003)
  • L. Yang et al.

    Abrogation of TGF beta signaling in mammary carcinomas recruits Gr-1+ CD11b+ myeloid cells that promote metastasis

    Cancer Cell

    (2008)
  • D. Padua et al.

    TGFbeta primes breast tumors for lung metastasis seeding through angiopoietin-like 4

    Cell

    (2008)
  • L. Gorelik et al.

    Abrogation of TGFbeta signaling in T cells leads to spontaneous T cell differentiation and autoimmune disease

    Immunity

    (2000)
  • J.H. Kehrl et al.

    Production of transforming growth factor beta by human T lymphocytes and its potential role in the regulation of T cell growth

    J Exp Med

    (1986)
  • J.H. Kehrl et al.

    Transforming growth factor beta is an important immunomodulatory protein for human B lymphocytes

    J Immunol

    (1986)
  • A.H. Rook et al.

    Effects of transforming growth factor beta on the functions of natural killer cells: depressed cytolytic activity and blunting of interferon responsiveness

    J Immunol

    (1986)
  • A. Fontana et al.

    Expression of TGF-beta 2 in human glioblastoma: a role in resistance to immune rejection?

    Ciba Found Symp

    (1991)
  • J.J. Mule et al.

    Transforming growth factor-beta inhibits the in vitro generation of lymphokine-activated killer cells and cytotoxic T cells

    Cancer Immunol Immunother

    (1988)
  • L.M. Coussens et al.

    Inflammation and cancer

    Nature

    (2002)
  • K.E. de Visser et al.

    Paradoxical roles of the immune system during cancer development

    Nat Rev Cancer

    (2006)
  • M.O. Li et al.

    Transforming growth factor-beta regulation of immune responses

    Annu Rev Immunol

    (2006)
  • S.H. Wrzesinski et al.

    Transforming growth factor-beta and the immune response: implications for anticancer therapy

    Clin Cancer Res

    (2007)
  • J.J. Chen et al.

    Regulation of the proinflammatory effects of Fas ligand (CD95L)

    Science

    (1998)
  • B. Bierie et al.

    Tumour microenvironment: TGFbeta: the molecular Jekyll and Hyde of cancer

    Nat Rev Cancer

    (2006)
  • R. Derynck et al.

    Synthesis of messenger RNAs for transforming growth factors alpha and beta and the epidermal growth factor receptor by human tumors

    Cancer Res

    (1987)
  • R.B. Dickson et al.

    Activation of growth factor secretion in tumorigenic states of breast cancer induced by 17 beta-estradiol or v-Ha-ras oncogene

    Proc Natl Acad Sci USA

    (1987)
  • S. Wojtowicz-Praga

    Reversal of tumor-induced immunosuppression by TGF-beta inhibitors

    Invest New Drugs

    (2003)
  • D.G. Stover et al.

    A delicate balance: TGF-beta and the tumor microenvironment

    J Cell Biochem

    (2007)
  • M. Abe et al.

    Cell-associated activation of latent transforming growth factor-beta by calpain

    J Cell Physiol

    (1998)
  • R.M. Lyons et al.

    Proteolytic activation of latent transforming growth factor-beta from fibroblast-conditioned medium

    J Cell Biol

    (1988)
  • K. Miyazono et al.

    Role for carbohydrate structures in TGF-beta 1 latency

    Nature

    (1989)
  • Q. Yu et al.

    Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-beta and promotes tumor invasion and angiogenesis

    Genes Dev

    (2000)
  • S. Schultz-Cherry et al.

    Influenza virus neuraminidase activates latent transforming growth factor beta

    J Virol

    (1996)
  • S. Schultz-Cherry et al.

    Thrombospondin causes activation of latent transforming growth factor-beta secreted by endothelial cells by a novel mechanism

    J Cell Biol

    (1993)
  • M.H. Barcellos-Hoff et al.

    Redox-mediated activation of latent transforming growth factor-beta 1

    Mol Endocrinol

    (1996)
  • R. Derynck et al.

    Smad-dependent and Smad-independent pathways in TGF-beta family signalling

    Nature

    (2003)
  • M.K. Lee et al.

    TGF-beta activates Erk MAP kinase signalling through direct phosphorylation of ShcA

    EMBO J

    (2007)
  • X.H. Feng et al.

    Specificity and versatility in TGF-signaling through Smads

    Annu Rev Cell Dev Biol

    (2005)
  • A.C. Daly et al.

    Transforming growth factor beta-induced Smad1/5 phosphorylation in epithelial cells is mediated by novel receptor complexes and is essential for anchorage-independent growth

    Mol Cell Biol

    (2008)
  • A. Moustakas et al.

    Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression

    Cancer Sci

    (2007)
  • Q. Zeng et al.

    Tgfbr1 haploinsufficiency is a potent modifier of colorectal cancer development

    Cancer Res

    (2009)
  • L. Valle et al.

    Germline allele-specific expression of TGFBR1 confers an increased risk of colorectal cancer

    Science

    (2008)
  • Cited by (0)

    Brian Bierie, Ph.D. performed his undergraduate work at Colorado State University in his home town of Fort Collins, Colorado. After finishing his undergraduate work he spent time working in the area of analytical chemistry for a large pesticide production plant. During this time he realized that some of the mutagens were so abundant in the soil and waste water produced by the production facilities that he began to think about dedicating his time to studies that could help people rather than contributing to the production of industrial mutagenic compounds. As a result, he switched to the study mammary gland biology and neoplasia mentored by Dr. Lothar Hennighausen, Ph.D. at the NIH in Bethesda, Maryland. After a three year tour at NIH he initiated his Ph.D. studies mentored by Dr. Harold L. Moses, M.D. at Vanderbilt University in Nashville, Tennessee. He received his Ph.D. in Cancer Biology and is now working as a postdoc mentored by Dr. Robert Weinberg, Ph.D. at the Whitehead Institute for Biomedical Research in Boston, Massachusetts.

    Harold L. Moses, M.D. is the Hortense B. Ingram Professor of Molecular Oncology and Professor of Cancer Biology, Medicine and Pathology at Vanderbilt University School of Medicine. He was the founding director of the Vanderbilt-Ingram Cancer Center, which he led for twelve years; he is now director emeritus. Trained as a pathologist, he has devoted much of his career to basic research on growth factors and tumor suppressor genes, particularly in the TGF-β pathway, and has received many awards for his research. He has served as president of the American Association for Cancer Research, president of the Association of American Cancer Institutes, chair of the NIH Chemical Pathology Study Section, chair of the Molecular Oncogenesis Study Section, and chair of the National Cancer Institute Cancer Centers review panel. He is a member of the Institute of Medicine of the National Academies and currently chairs the National Cancer Policy Forum of the Institute of Medicine. He also currently co-chairs the Program Steering Committee for the NCI's Tumor Microenvironment Network.

    View full text