Skip to main content
Top
Published in: Cancer Microenvironment 1/2012

01-04-2012 | Review Paper

Epithelial-Mesenchymal Transition Induced by Senescent Fibroblasts

Authors: Remi-Martin Laberge, Pierre Awad, Judith Campisi, Pierre-Yves Desprez

Published in: Cancer Microenvironment | Issue 1/2012

Login to get access

Abstract

Depending on the cell type and tissue environment, epithelial and mesenchymal cell phenotypes are not static and can be highly dynamic. Epithelial-mesenchymal transitions (EMTs) and reverse EMTs provide flexibility during embryogenesis. While EMTs are a critical normal process during development and wound healing, properties of the EMT have been implicated in human pathology, particularly cancer metastasis. A normal undamaged epithelium does not typically exhibit features of an EMT. However, particularly under the influence of the surrounding microenvironment, cancer cells may reactivate developmental phenotypes out of context in the adult. This reactivation, such as the EMT, can facilitate tumor cell invasion and metastasis, and therefore is a major mechanism of tumor progression. Conversely, cellular senescence, which is associated with aging, is a process by which cells enter a state of permanent cell cycle arrest, thereby constituting a potent tumor suppressive mechanism. However, accumulating evidence shows that senescent cells can have deleterious effects on the tissue microenvironment. The most significant of these effects is the acquisition of a senescence-associated secretory phenotype (SASP) that turns senescent fibroblasts into pro-inflammatory cells having the ability to promote tumor progression, in part by inducing an EMT in nearby epithelial cells. Here, we summarize the potential impacts of SASP factors, particularly interleukins, on tissue microenvironments and their ability to stimulate tumor progression through induction of an EMT.
Literature
1.
go back to reference Kalluri R, Weinberg RA (2009) The basics of epithelial-mesenchymal transition. J Clin Invest 119(6):1420–1428PubMedCrossRef Kalluri R, Weinberg RA (2009) The basics of epithelial-mesenchymal transition. J Clin Invest 119(6):1420–1428PubMedCrossRef
2.
go back to reference Micalizzi DS, Farabaugh SM, Ford HL (2010) Epithelial-mesenchymal transition in cancer: parallels between normal development and tumor progression. J Mammary Gland Biol Neoplasia 15(2):117–134PubMedCrossRef Micalizzi DS, Farabaugh SM, Ford HL (2010) Epithelial-mesenchymal transition in cancer: parallels between normal development and tumor progression. J Mammary Gland Biol Neoplasia 15(2):117–134PubMedCrossRef
3.
go back to reference Peinado H, Portillo F, Cano A (2004) Transcriptional regulation of cadherins during development and carcinogenesis. Int J Dev Biol 48(5–6):365–375PubMedCrossRef Peinado H, Portillo F, Cano A (2004) Transcriptional regulation of cadherins during development and carcinogenesis. Int J Dev Biol 48(5–6):365–375PubMedCrossRef
4.
go back to reference Birchmeier W, Behrens J (1994) Cadherin expression in carcinomas: role in the formation of cell junctions and the prevention of invasiveness. Biochim Biophys Acta 1198(1):11–26PubMed Birchmeier W, Behrens J (1994) Cadherin expression in carcinomas: role in the formation of cell junctions and the prevention of invasiveness. Biochim Biophys Acta 1198(1):11–26PubMed
5.
go back to reference Prasad CP et al (2009) Expression analysis of E-cadherin, Slug and GSK3beta in invasive ductal carcinoma of breast. BMC Cancer 9:325PubMedCrossRef Prasad CP et al (2009) Expression analysis of E-cadherin, Slug and GSK3beta in invasive ductal carcinoma of breast. BMC Cancer 9:325PubMedCrossRef
6.
go back to reference Logullo AF et al (2010) Concomitant expression of epithelial-mesenchymal transition biomarkers in breast ductal carcinoma: association with progression. Oncol Rep 23(2):313–320PubMed Logullo AF et al (2010) Concomitant expression of epithelial-mesenchymal transition biomarkers in breast ductal carcinoma: association with progression. Oncol Rep 23(2):313–320PubMed
7.
go back to reference Mori M et al (2009) Zyxin mediates actin fiber reorganization in epithelial-mesenchymal transition and contributes to endocardial morphogenesis. Mol Biol Cell 20(13):3115–3124PubMedCrossRef Mori M et al (2009) Zyxin mediates actin fiber reorganization in epithelial-mesenchymal transition and contributes to endocardial morphogenesis. Mol Biol Cell 20(13):3115–3124PubMedCrossRef
8.
go back to reference Orimo A, Weinberg RA (2006) Stromal fibroblasts in cancer: a novel tumor-promoting cell type. Cell Cycle 5(15):1597–1601PubMedCrossRef Orimo A, Weinberg RA (2006) Stromal fibroblasts in cancer: a novel tumor-promoting cell type. Cell Cycle 5(15):1597–1601PubMedCrossRef
9.
go back to reference Olumi AF et al (1999) Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res 59:5002–5011PubMed Olumi AF et al (1999) Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res 59:5002–5011PubMed
10.
go back to reference Erez N et al (2010) Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell 17(2):135–147PubMedCrossRef Erez N et al (2010) Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell 17(2):135–147PubMedCrossRef
11.
go back to reference Qiu W et al (2008) No evidence of clonal somatic genetic alterations in cancer-associated fibroblasts from human breast and ovarian carcinomas. Nat Genet 40(5):650–655PubMedCrossRef Qiu W et al (2008) No evidence of clonal somatic genetic alterations in cancer-associated fibroblasts from human breast and ovarian carcinomas. Nat Genet 40(5):650–655PubMedCrossRef
12.
go back to reference Rodier F et al (2009) Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol 11(8):973–979PubMedCrossRef Rodier F et al (2009) Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol 11(8):973–979PubMedCrossRef
13.
go back to reference Gilbert LA, Hemann MT (2010) DNA damage-mediated induction of a chemoresistant niche. Cell 143(3):355–366PubMedCrossRef Gilbert LA, Hemann MT (2010) DNA damage-mediated induction of a chemoresistant niche. Cell 143(3):355–366PubMedCrossRef
14.
go back to reference Hayflick L (1965) The limited in vitro lifetime of human diploid cell strains. Exp Cell Res 37:614–636PubMedCrossRef Hayflick L (1965) The limited in vitro lifetime of human diploid cell strains. Exp Cell Res 37:614–636PubMedCrossRef
15.
go back to reference d’Adda di Fagagna F et al (2003) A DNA damage checkpoint response in telomere-initiated senescence. Nature 426:194–198PubMedCrossRef d’Adda di Fagagna F et al (2003) A DNA damage checkpoint response in telomere-initiated senescence. Nature 426:194–198PubMedCrossRef
16.
go back to reference Campisi J, d’Adda di Fagagna F (2007) Cellular senescence: when bad things happen to good cells. Nature Rev Molec Cell Biol 8:729–740CrossRef Campisi J, d’Adda di Fagagna F (2007) Cellular senescence: when bad things happen to good cells. Nature Rev Molec Cell Biol 8:729–740CrossRef
17.
go back to reference Gorgoulis VG, Halazonetis TD (2010) Oncogene-induced senescence: the bright and dark side of the response. Curr Opin Cell Biol 22(6):816–827PubMedCrossRef Gorgoulis VG, Halazonetis TD (2010) Oncogene-induced senescence: the bright and dark side of the response. Curr Opin Cell Biol 22(6):816–827PubMedCrossRef
18.
go back to reference Prieur A, Peeper DS (2008) Cellular senescence in vivo: a barrier to tumorigenesis. Curr Opin Cell Biol 20:150–155PubMedCrossRef Prieur A, Peeper DS (2008) Cellular senescence in vivo: a barrier to tumorigenesis. Curr Opin Cell Biol 20:150–155PubMedCrossRef
20.
go back to reference Coppe JP et al (2010) The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol 5:99–118PubMedCrossRef Coppe JP et al (2010) The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol 5:99–118PubMedCrossRef
21.
go back to reference Coppe JP et al (2008) Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol 6(12):2853–2868PubMedCrossRef Coppe JP et al (2008) Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol 6(12):2853–2868PubMedCrossRef
22.
go back to reference Campisi J (2005) Senescent cells, tumor suppression and organismal aging: good citizens, bad neighbors. Cell 120:513–522PubMedCrossRef Campisi J (2005) Senescent cells, tumor suppression and organismal aging: good citizens, bad neighbors. Cell 120:513–522PubMedCrossRef
23.
go back to reference Dimri GP et al (1995) A novel biomarker identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci USA 92:9363–9367PubMedCrossRef Dimri GP et al (1995) A novel biomarker identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci USA 92:9363–9367PubMedCrossRef
24.
go back to reference Jeyapalan JC et al (2007) Accumulation of senescent cells in mitotic tissue of aging primates. Mech Ageing Dev 128:36–44PubMedCrossRef Jeyapalan JC et al (2007) Accumulation of senescent cells in mitotic tissue of aging primates. Mech Ageing Dev 128:36–44PubMedCrossRef
25.
go back to reference Paradis V et al (2001) Replicative senescence in normal liver, chronic hepatitis C, and hepatocellular carcinomas. Hum Pathol 32:327–332PubMedCrossRef Paradis V et al (2001) Replicative senescence in normal liver, chronic hepatitis C, and hepatocellular carcinomas. Hum Pathol 32:327–332PubMedCrossRef
26.
go back to reference Erusalimsky JD, Kurz DJ (2005) Cellular senescence in vivo: its relevance in ageing and cardiovascular disease. Exp Gerontol 40(8–9):634–642PubMedCrossRef Erusalimsky JD, Kurz DJ (2005) Cellular senescence in vivo: its relevance in ageing and cardiovascular disease. Exp Gerontol 40(8–9):634–642PubMedCrossRef
27.
go back to reference Martin JA, Buckwalter JA (2003) The role of chondrocyte senescence in the pathogenesis of osteoarthritis and in limiting cartilage repair. J Bone Joint Surg Am 85:106–110PubMed Martin JA, Buckwalter JA (2003) The role of chondrocyte senescence in the pathogenesis of osteoarthritis and in limiting cartilage repair. J Bone Joint Surg Am 85:106–110PubMed
28.
go back to reference Roberts S et al (2006) Senescence in human intervertebral discs. Eur Spine J 15:312–316CrossRef Roberts S et al (2006) Senescence in human intervertebral discs. Eur Spine J 15:312–316CrossRef
29.
go back to reference Finch CE, Crimmins EM (2004) Inflammatory exposure and historical changes in human life-spans. Science 305:1736–1739PubMedCrossRef Finch CE, Crimmins EM (2004) Inflammatory exposure and historical changes in human life-spans. Science 305:1736–1739PubMedCrossRef
30.
go back to reference Parrinello S et al (2005) Stromal-epithelial interactions in aging and cancer: senescent fibroblasts alter epithelial cell differentiation. J Cell Sci 118(Pt 3):485–496PubMedCrossRef Parrinello S et al (2005) Stromal-epithelial interactions in aging and cancer: senescent fibroblasts alter epithelial cell differentiation. J Cell Sci 118(Pt 3):485–496PubMedCrossRef
31.
go back to reference Freund A et al (2010) Inflammatory networks during cellular senescence: causes and consequences. Trends Mol Med 16(5):238–246PubMedCrossRef Freund A et al (2010) Inflammatory networks during cellular senescence: causes and consequences. Trends Mol Med 16(5):238–246PubMedCrossRef
33.
go back to reference Krtolica A et al (2001) Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging. Proc Natl Acad Sci USA 98:12072–12077PubMedCrossRef Krtolica A et al (2001) Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging. Proc Natl Acad Sci USA 98:12072–12077PubMedCrossRef
34.
go back to reference Liu D, Hornsby PJ (2007) Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion. Cancer Res 67:3117–3126PubMedCrossRef Liu D, Hornsby PJ (2007) Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion. Cancer Res 67:3117–3126PubMedCrossRef
35.
go back to reference Coppe JP et al (2010) A human-like senescence-associated secretory phenotype is conserved in mouse cells dependent on physiological oxygen. PLoS One 5(2):e9188PubMedCrossRef Coppe JP et al (2010) A human-like senescence-associated secretory phenotype is conserved in mouse cells dependent on physiological oxygen. PLoS One 5(2):e9188PubMedCrossRef
36.
go back to reference Bavik C et al (2006) The gene expression program of prostate fibroblast senescence modulates neoplastic epithelial cell proliferation through paracrine mechanisms. Cancer Res 66:794–802PubMedCrossRef Bavik C et al (2006) The gene expression program of prostate fibroblast senescence modulates neoplastic epithelial cell proliferation through paracrine mechanisms. Cancer Res 66:794–802PubMedCrossRef
37.
go back to reference Wang B et al (2006) A growth-related oncogene/CXC chemokine receptor 2 autocrine loop contributes to cellular proliferation in esophageal cancer. Cancer Res 66:3071–3077PubMedCrossRef Wang B et al (2006) A growth-related oncogene/CXC chemokine receptor 2 autocrine loop contributes to cellular proliferation in esophageal cancer. Cancer Res 66:3071–3077PubMedCrossRef
38.
go back to reference Tsai KK et al (2005) Cellular mechanisms for low-dose ionizing radiation-induced perturbation of the breast tissue microenvironment. Cancer Res 65:6734–6744PubMedCrossRef Tsai KK et al (2005) Cellular mechanisms for low-dose ionizing radiation-induced perturbation of the breast tissue microenvironment. Cancer Res 65:6734–6744PubMedCrossRef
39.
go back to reference Ohuchida K et al (2004) Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor-stromal interactions. Cancer Res 64(9):3215–3222PubMedCrossRef Ohuchida K et al (2004) Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor-stromal interactions. Cancer Res 64(9):3215–3222PubMedCrossRef
40.
go back to reference Potempa S, Ridley AJ (1998) Activation of both MAP kinase and phosphatidylinositide 3-kinase by Ras is required for hepatocyte growth factor/scatter factor-induced adherens junction disassembly. Mol Biol Cell 9(8):2185–2200PubMed Potempa S, Ridley AJ (1998) Activation of both MAP kinase and phosphatidylinositide 3-kinase by Ras is required for hepatocyte growth factor/scatter factor-induced adherens junction disassembly. Mol Biol Cell 9(8):2185–2200PubMed
41.
go back to reference Paumelle R et al (2002) Hepatocyte growth factor/scatter factor activates the ETS1 transcription factor by a RAS-RAF-MEK-ERK signaling pathway. Oncogene 21(15):2309–2319PubMedCrossRef Paumelle R et al (2002) Hepatocyte growth factor/scatter factor activates the ETS1 transcription factor by a RAS-RAF-MEK-ERK signaling pathway. Oncogene 21(15):2309–2319PubMedCrossRef
42.
go back to reference Thiery JP (2002) Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2(6):442–454PubMedCrossRef Thiery JP (2002) Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2(6):442–454PubMedCrossRef
43.
go back to reference Tonini T, Rossi F, Claudio PP (2003) Molecular basis of angiogenesis and cancer. Oncogene 22(42):6549–6556PubMedCrossRef Tonini T, Rossi F, Claudio PP (2003) Molecular basis of angiogenesis and cancer. Oncogene 22(42):6549–6556PubMedCrossRef
44.
go back to reference Birchmeier C et al (2003) Met, metastasis, motility and more. Nat Rev Mol Cell Biol 4(12):915–925PubMedCrossRef Birchmeier C et al (2003) Met, metastasis, motility and more. Nat Rev Mol Cell Biol 4(12):915–925PubMedCrossRef
45.
go back to reference Yuan A et al (2005) The role of interleukin-8 in cancer cells and microenvironment interaction. Front Biosci 10:853–865PubMedCrossRef Yuan A et al (2005) The role of interleukin-8 in cancer cells and microenvironment interaction. Front Biosci 10:853–865PubMedCrossRef
46.
go back to reference Badache A, Hynes NE (2001) Interleukin 6 inhibits proliferation and, in cooperation with an epidermal growth factor receptor autocrine loop, increases migration of T47D breast cancer cells. Cancer Res 61:383–391PubMed Badache A, Hynes NE (2001) Interleukin 6 inhibits proliferation and, in cooperation with an epidermal growth factor receptor autocrine loop, increases migration of T47D breast cancer cells. Cancer Res 61:383–391PubMed
47.
go back to reference Camphausen K et al (2001) Radiation therapy to a primary tumor accelerates metastatic growth in mice. Cancer Res 61(5):2207–2211PubMed Camphausen K et al (2001) Radiation therapy to a primary tumor accelerates metastatic growth in mice. Cancer Res 61(5):2207–2211PubMed
48.
go back to reference Qian LW et al (2002) Radiation-induced increase in invasive potential of human pancreatic cancer cells and its blockade by a matrix metalloproteinase inhibitor, CGS27023. Clin Cancer Res 8(4):1223–1227PubMed Qian LW et al (2002) Radiation-induced increase in invasive potential of human pancreatic cancer cells and its blockade by a matrix metalloproteinase inhibitor, CGS27023. Clin Cancer Res 8(4):1223–1227PubMed
49.
go back to reference Coppe JP et al (2008) A role for fibroblasts in mediating the effects of tobacco-induced epithelial cell growth and invasion. Mol Cancer Res 6(7):1085–1098PubMedCrossRef Coppe JP et al (2008) A role for fibroblasts in mediating the effects of tobacco-induced epithelial cell growth and invasion. Mol Cancer Res 6(7):1085–1098PubMedCrossRef
50.
go back to reference Coppe JP et al (2006) Secretion of vascular endothelial growth factor by primary human fibroblasts at senescence. J Biol Chem 281(40):29568–29574PubMedCrossRef Coppe JP et al (2006) Secretion of vascular endothelial growth factor by primary human fibroblasts at senescence. J Biol Chem 281(40):29568–29574PubMedCrossRef
51.
go back to reference Strieter RM et al (2006) Cancer CXC chemokine networks and tumour angiogenesis. Eur J Cancer 42(6):768–778PubMedCrossRef Strieter RM et al (2006) Cancer CXC chemokine networks and tumour angiogenesis. Eur J Cancer 42(6):768–778PubMedCrossRef
52.
go back to reference Orr FW, Wang HH (2001) Tumor cell interactions with the microvasculature: a rate-limiting step in metastasis. Surg Oncol Clin N Am 10(2):357–381, ix-xPubMed Orr FW, Wang HH (2001) Tumor cell interactions with the microvasculature: a rate-limiting step in metastasis. Surg Oncol Clin N Am 10(2):357–381, ix-xPubMed
53.
go back to reference Nickoloff BJ et al (2004) Tumor suppressor maspin is up-regulated during keratinocyte senescence, exerting a paracrine antiangiogenic activity. Cancer Res 64(9):2956–2961PubMedCrossRef Nickoloff BJ et al (2004) Tumor suppressor maspin is up-regulated during keratinocyte senescence, exerting a paracrine antiangiogenic activity. Cancer Res 64(9):2956–2961PubMedCrossRef
54.
55.
go back to reference Xue W et al (2007) Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 445:656–650PubMedCrossRef Xue W et al (2007) Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 445:656–650PubMedCrossRef
56.
go back to reference Homey B, Muller A, Zlotnik A (2002) Chemokines: agents for the immunotherapy of cancer? Nat Rev Immunol 2(3):175–184PubMedCrossRef Homey B, Muller A, Zlotnik A (2002) Chemokines: agents for the immunotherapy of cancer? Nat Rev Immunol 2(3):175–184PubMedCrossRef
58.
go back to reference Ben-Baruch A (2006) Inflammation-associated immune suppression in cancer: the roles played by cytokines, chemokines and additional mediators. Semin Cancer Biol 16(1):38–52PubMedCrossRef Ben-Baruch A (2006) Inflammation-associated immune suppression in cancer: the roles played by cytokines, chemokines and additional mediators. Semin Cancer Biol 16(1):38–52PubMedCrossRef
59.
go back to reference Cowin P, Rowlands TM, Hatsell SJ (2005) Cadherins and catenins in breast cancer. Curr Opin Cell Biol 17:499–508PubMedCrossRef Cowin P, Rowlands TM, Hatsell SJ (2005) Cadherins and catenins in breast cancer. Curr Opin Cell Biol 17:499–508PubMedCrossRef
60.
go back to reference Kokkinos MI et al (2007) Vimentin and epithelial-mesenchymal transition in human breast cancer—observations in vitro and in vivo. Cells Tissues Organs 185:191–203PubMedCrossRef Kokkinos MI et al (2007) Vimentin and epithelial-mesenchymal transition in human breast cancer—observations in vitro and in vivo. Cells Tissues Organs 185:191–203PubMedCrossRef
61.
go back to reference Dhawan P et al (2005) Claudin-1 regulates cellular transformation and metastatic behavior in colon cancer. J Clin Invest 115:1765–1776PubMedCrossRef Dhawan P et al (2005) Claudin-1 regulates cellular transformation and metastatic behavior in colon cancer. J Clin Invest 115:1765–1776PubMedCrossRef
63.
go back to reference Bissell MJ, Radisky D (2001) Putting tumours in context. Nature Rev Cancer 1:46–54CrossRef Bissell MJ, Radisky D (2001) Putting tumours in context. Nature Rev Cancer 1:46–54CrossRef
65.
go back to reference Brabletz T et al (2005) Invasion and metastasis in colorectal cancer: epithelial-mesenchymal transition, mesenchymal-epithelial transition, stem cells and beta-catenin. Cells Tissues Organs 179(1–2):56–65PubMedCrossRef Brabletz T et al (2005) Invasion and metastasis in colorectal cancer: epithelial-mesenchymal transition, mesenchymal-epithelial transition, stem cells and beta-catenin. Cells Tissues Organs 179(1–2):56–65PubMedCrossRef
Metadata
Title
Epithelial-Mesenchymal Transition Induced by Senescent Fibroblasts
Authors
Remi-Martin Laberge
Pierre Awad
Judith Campisi
Pierre-Yves Desprez
Publication date
01-04-2012
Publisher
Springer Netherlands
Published in
Cancer Microenvironment / Issue 1/2012
Print ISSN: 1875-2292
Electronic ISSN: 1875-2284
DOI
https://doi.org/10.1007/s12307-011-0069-4

Other articles of this Issue 1/2012

Cancer Microenvironment 1/2012 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine