Abstract
Cancers become significantly more dangerous when the tumor progresses from in situ, or contained, to an invasive state, in which the cancer cells acquire the ability to pass through the surrounding basement membrane (BM), a specialized extracellular matrix (ECM) that provides structure and contextual information to the underlying tissue. While the majority of tumors are carcinomas, derived from epithelial cells, it is the stromal cells surrounding the epithelial-derived tumor cells, including fibroblasts and myofibroblasts, vasculature, and immune cells, that are largely responsible for the production and remodeling of the ECM. Here, we will discuss myofibroblasts as key effectors of tumor progression, focusing on recent advances in breast and pancreatic carcinoma, showing how myofibroblasts may function properly in normal tissue remodeling and wound-healing processes, how in the tumor context they can drive cancer invasion and metastasis, and how the pathogenic functions of myofibroblasts may be targeted therapeutically.
Keywords: Myofibroblast, breast cancer, pancreatic cancer, epithelial-mesenchymal transition, extracellular matrix, tissue disruption
Introduction and context
Understanding the key factors in development and progression of a particular cancer type is critical for identifying therapeutic interventions that target cancer at the earliest stages of development. Environmental, nutritional, and hereditary factors that have been identified as associated with increased cancer risk could potentially reveal those processes essential for cancer development. Surprisingly, while cancer risk prediction models that incorporate such risk factors have been found to perform well at the population level, they have not translated as well to individualized cancer risk assessment [1]. Such findings have led to the realization that apart from the cancer cells, the composition and structure of the ECM plays an important inhibitory role in cancer development [2]. During a person's lifetime, individual cells may accumulate potentially tumorigenic genetic mutations, but only rarely do these develop into cancer. Cancer development might be viewed as a landslide, where somatic mutations can be viewed as pre-existing stresses, and exposure to carcinogenic toxins as a continuous rain. These mutations may lead to development of abnormalities, and even to carcinoma in situ, but so long as the underlying ECM remains intact, tumor development is slowed. Once myofibroblasts become activated, however the ECM becomes damaged and degraded; the former solid surface becomes loose, leading to system collapses, systemic disease, and death.
The immune system can detect the existence of some oncogenic alterations, in a similar fashion to identification of viruses and bacteria. Initially, the immune system might attempt to “heal” the cancer in the same way it heals wounds, invading the tissue and transforming the phenotypes of resident cells [3]. What works well in wounds, however, can go awry in the innate fight against cancer, where the roles of myofibroblasts and even their origins are not entirely parallel. One major difference between wound myofibroblasts and tumor associated myofibroblasts is the ability of the latter to influence epithelial cells to proliferate and to drive malignant transformation [4]. Another difference is that in wounds, myofibroblasts appear to be differentiated from mesenchymal cells, while in the tumor microenvironment, myofibroblasts can develop directly from the epithelial cells as well as from the mesenchymal cells [5, 6].
In normal wound healing, the initiating damage causes the activation of the coagulation cascade that in turn activates the innate immune response. With the arrival of myeloid-derived immune cells, the signal for cytokine production leads towards activation of the adaptive immune system. Secreted cytokines activate fibroblasts and endothelial cells, which transform into myofibroblasts that stimulate angiogenesis, increase ECM production, and physically contract the wound edges, facilitating the healing process. The initiating factors including key cytokine Transforming Growth Factor-β (TGF-β) are eventually depleted as the wound heals, at which time the myofibroblasts disappear through apoptosis [3, 5]. If myofibroblasts persist, they transform the site into a chronic wound; examples of this phenomenon include skin keloids [7] and fibrosis of organs such as liver, lung or pancreas [8].
In cancer development, myofibroblasts are present in the stroma of most epithelial tumor types [9, 10] , where they can act as drivers of malignant transformation. The tumor-associated myofibroblasts express matrix metalloproteinases (MMPs) which can degrade nearly every component of the ECM, including the BM; myofibroblasts also possess the ability to contract the ECM, which disrupts normal epithelial tissue structures and provides pathways for tumor cells to invade [11]. Secreted MMPs interact directly with the carcinoma cells, breaking down cell-cell junctions and adhesions, further facilitating tumor cell movement [12]. MMPs can also directly stimulate tumor progression through promotion of the epithelial mesenchymal transition (EMT) [13], a developmental process that activates a migratory and invasive cellular phenotype in epithelial tumor cells and that can under chronic activation conditions cause epithelial cells to transition entirely into a myofibroblast phenotype [14].
Fibrosis and myofibroblasts
Myofibroblasts are ubiquitously found in the human body, in the skin during wound healing [8, 15], the heart [16, 17] and the intestines [4]. They are detectable by week 21 during embryogenesis [4] while in adults, stellate cells in liver, pancreas, and lung possess myofibroblast charateristics. During activation, myofibroblasts develop actin stress fibers and express α-smooth muscle actin (αSMA) [3, 15, 18, 19]. In 1972 Gabbiani et al. described fibroblasts in their ability to become contractile cells by undergoing essential changes of their inner architecture as well as their membrane structure [20]. In the years after the first description this cell type was found in all types of connective and soft tissue disorders such as fibrosarcoma [21], arterial dysplasia [22] and also in organ specific parenchymal disorders such as alcoholic cirrhosis [23].
Fibrosis is defined as the disproportionate increase of fibrous connective tissue, collagens, and fibronectin [19]. Usually occurring first around inflamed or injured tissue, fibrosis can be found in bening skin conditions such as Dupuytren's contracture [24], and can be a life-threatening disease when it affects the internal organs such as lung, liver, kidneys, and pancreas [8]. Myofibroblasts are the key players in the development of fibrosis through their ability to produce large amounts of ECM and to modulate contraction, leading to a very dense tissue appearance as seen for example in fibrosis of the liver, lung, or breast [5, 25–27]. Expression of TGF-β is central to fibrosis generation and maintenance. The three isoforms of TGF-β(β1, β2, β3) bind to TGF-β receptor II (TGF-βRII) which will then generate a complex with TGFβRI; this heteromeric receptor complex then phosphorylates Smad transcription factors which in turn translocate into the nucleus to control transcription [28–30]. TGF-β1 has been the best studied of the isoforms for its role in activation and signaling pathways of myofibroblasts [31]
TGF-β1 can induce myofibroblast differentiation from fibroblasts; inhibiting TGF-β1 signaling in fibrosis leads to reduced expression of myofibroblast markers and decreased tissue contraction [15, 16, 32]. The activating TGF-β1 can derive from invading immune cells, epithelial cells, or through fibroblast autocrine pathways [33]. Other cytokines such as TNF-α have been shown to facilitate TGF-β induced myofibroblast activation and fibrosis promotion [34]. In addition reactive oxygen species (ROS) have also been implicated in TGF-β-induced myofibroblast formation, and the antioxidant N-acetylcysteine has been assessed as a potential antifibrosis therapeutic [35]. The mechanical and oxidative stress that leads to the production of ROS can alter the gene expression in fibroblasts as well [36, 37] .
Cancer and myofibroblasts
Cancer-associated myofibroblasts possess many of the features found in nonmalignant tissue. However, there is an important difference: in normal wound healing, elevated levels of TGF-β1 in the wound microenvironment has a powerful cytostatic effect on epithelial cells, but once the epithelial cells have acquired specific mutations, the cytostatic effect of TGF-β is lost [38, 39]. Additionally, the metaplastic characteristic of tumors allows for a wider range of cellular differentiation processes, and many instances of epithelial-derived, cancer-associated myofibroblasts have been found. In many cases, these may differentiate from the cancer cells themselves through EMT, which is facilitated both by the cytokines produced by cancer cells, the invading immune cells [40, 41], and by the elevated levels of ROS found in tumors [42]. ROS can stimulate the expression of Snail, which can activate the EMT program, including downregulation of E-cadherin, increased invasiveness, and expression of mesenchymal markers [43–45]. TGF-β1 can stimulate ROS production in endothelial cells as well, triggering conversion to myofibroblasts through autocrine and paracrine effects [46]. ROS also directly activate fibroblasts, producing myofibroblasts with mitochondrial dysfunction and alterations in aerobic glycolysis that increase production of l-lactate, ketone bodies, and glutamine; these nutrients function as energy sources for the cancer cells’ anabolic growth and oxidative mitochondrial metabolism [47–50].
Breast cancer
Myofibroblasts are found in all subtypes of breast cancer [12, 51, 52], and are a major component of the breast cancer mass. Myofibroblast abundance correlates with lymph node metastasis, higher clinical grade, and poor overall survival prognosis [53]. Most breast cancer cell lines can be induced to undergo EMT, but ongoing activation of EMT is particularly evident in metaplastic breast carcinomas, a breast cancer subtype with particularly poor prognosis [54]. Direct experimental evidence for generation of myofibroblasts from tumor cells came from experiments in which fibroblast cell lines were derived from a metaplastic breast tumor [6]. These cell lines were found to have common patterns of X-chromosome inactivation with the parental tumor, indicating a common cellular origin, but were found to express mesenchymal markers. Furthermore, the lines behaved in vitro and in vivo as normal fibroblasts, but when combined with tumor cells, expressed αSMA and acquired the phenotypic appearance of myofibroblasts. Strikingly, the tumor-derived fibroblasts were nonmalignant when injected into immunocompromised mice, but substantially enhanced tumor growth when coinjected with MCF7 breast cancer cells [6]. MMPs, abundant in breast cancer, can also directly activate myofibroblast characteristics in nonmalignant mammary epithelial cells through a pathway involving increased expression of the splice isoform Rac1b and increased production of ROS [43].
Some of the same processes that regulate tumor cell malignancy can also affect how myofibroblasts function. Examination of p16INK4A levels in multiple samples of tumor associated breast fibroblasts revealed that a significant proportion showed downregulation of protein expression [55]. p16INK4A is a cyclin dependent kinase inhibitor that regulates cell cycle progression and is expressed in cells undergoing senescence, an intrinsic antiproliferative mechanism. These experiments showed further, that reduced p16INK4a was associated with reduced expression of αSMA. Conditioned media from fibroblasts with downregulated p16INK4a stimulated proliferation of epithelial cells in vitro and when coinjected with MDAMB231 cells in orthotopic xenografts lead to faster tumor progression [55]. Myofibroblasts and breast cancer cells can interact with each other in complex ways. During tumor invasion, TGF-β1 signaling causes “single strand migration” of myofibroblasts and tumor cells, while inhibition of TGF-β results in compact movement of the invasion front of the tumor [56]. As another example, breast cancer cells can induce laminin 332 and integrin β4 expression in myofibroblasts, enhancing anoikis resistance for single detaching cells, thus stabilizing the surrounding ECM for the tumor[57].
Pancreatic cancer
In the pancreas, myofibroblasts are derived principally from pancreatic stellate cells (PSC). These cells are also important in the formation of chronic pancreatitis, a common comorbidity of pancreatic cancer and known predisposing factor in cancer development. PSCs are needed under normal circumstances to organize the ECM and to maintain the structural integrity of the organ. With tissue injury (caused by excessive alcohol, toxins, intake of fatty acids, or hypoglycemia), PSCs become activated to myofibroblasts, with characteristic development of microfilaments, expression of αSMA, increased production of ECM and MMPs, and increased contractility [58]. Similarly, in adenocarcinoma αSMA is found in the typical fibrillar pattern in the myofibroblasts, and in regions with high amounts of desmoplasia; procollagen I is also detected in activated myofibroblasts [59] (Figure 1).
Figure 1. Myofibroblast localization identified by smooth muscle actin staining (αSMA) in pancreatic ductal carcinoma.
(A,B) Normal pancreatic tissue, hematoxylin and eosin (H&E) staining (A) shows normal acinar cell and duct formation; in healthy tissue αSMA staining appears mainly in endothelial cells with rare presence of αSMA staining in connective tissue (B). (C,D) Ductal pancreatic carcinoma, H&E staining (C) shows disrupted gland formation with malignant cells, and large amounts of tumor surrounding stroma; strong positive staining of αSMA (D) reveals abundant myofibroblasts.
Specific mechanisms for activating pancreatic cancer-associated myofibroblasts have recently been identified: expression of paladin by pancreatic cancer cells stimulates myofibroblast activation [11], and expression in pancreatic myofibroblasts of the adhesion molecule L1CAM causes increased migratory ability and chemoresistance in pancreatic cancer cells [60, 61]. Pancreatic cancer-associated myofibroblasts may also act to drive metastasis by directly seeding the metastatic bed, as revealed by identification of Y-chromosomal positive pancreatic tumor-derived myofibroblasts in female mice in distant metastases [62].
Current challenges, and future directions
The near ubiquitous co-occurrence of fibrosis with cancer in the pancreas [63], liver [19, 64], and kidney [45], and the fact that chronic fibrosis is a predisposing factor for cancer development in these and other organ systems [5, 8, 45, 65] points to a central role of myofibroblasts in tumor development. The recent identification of additional direct interactions between tumor cells and myofibroblasts that potentiate tumor malignancy further highlights the importance of these cells. Therapeutic approaches that blocked myofibroblast development or function could have substantial value for treatment of fibrosis or cancer. However, while myofibroblasts are easy to identify in the tumor context, they are very difficult to study in culture, as their phenotype is very fluid, and it is unclear how much tissue-specificity is retained when separated from the in vivo context. Markers of myofibroblast tissue specificity are needed, as are more sophisticated culture methods for differentiating essential functions of myofibroblasts in the normal context from those that occur in chronic fibrosis and tumors. Even existing anticancer therapies may be more effective if we can block the destructive degradative action of the pathologic myofibroblasts, halting the landslide.
Acknowledgments
We thank Evette Radisky for helpful conversations and editing. This work is supported by NCI grants CA122086, 132789 and by the Mayo Clinic Breast SPORE CA116201.
Abbreviations
- BM
Basement membrane
- ECM
Extracellular matrix
- MMPs
Matrix metalloproteinases
- TGF-ß
Transforming growth factor-β
- ROS
Reactive oxygen species
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Rockhill B, Spiegelman D, Byrne C, Hunter DJ, Colditz GA. Validation of the Gail et al. Model of Breast Cancer Risk Prediction and Implications for Chemoprevention. JNCI J Natl Cancer Inst. 2001;93:358–366. doi: 10.1093/jnci/93.5.358. [DOI] [PubMed] [Google Scholar]
- 2.Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat. Rev. Cancer. 2006;6:392–401. doi: 10.1038/nrc1877. [DOI] [PubMed] [Google Scholar]
- 3.Hinz B, Gabbiani G. Mechanisms of force generation and transmission by myofibroblasts. Current Opinion in Biotechnology. 2003;14:538–546. doi: 10.1016/j.copbio.2003.08.006. [DOI] [PubMed] [Google Scholar]
- 4.Pinchuk IV, Mifflin RC, Saada JI, Powell DW. Intestinal Mesenchymal Cells. Curr Gastroenterol Rep. 2010;12:310–318. doi: 10.1007/s11894-010-0135-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Radisky DC, Kenny PA, Bissell MJ. Fibrosis and cancer: Do myofibroblasts come also from epithelial cells via EMT? Journal of Cellular Biochemistry. 2007;101:830–839. doi: 10.1002/jcb.21186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Petersen OW, Nielsen HL, Gudjonsson T, Villadsen R, Rank F, Niebuhr E, Bissell MJ, Rønnov-Jessen L. Epithelial to mesenchymal transition in human breast cancer can provide a nonmalignant stroma. Am. J. Pathol. 2003;162:391–402. doi: 10.1016/S0002-9440(10)63834-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Al-Attar A, Mess S, Thomassen JM, Kauffman CL, Davison SP. Keloid Pathogenesis and Treatment. Plastic and Reconstructive Surgery. 2006;117:286–300. doi: 10.1097/01.prs.0000195073.73580.46. [DOI] [PubMed] [Google Scholar]
- 8.Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat. Med. 2012;18:1028–1040. doi: 10.1038/nm.2807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bhowmick NA, Neilson EG, Moses HL. Stromal fibroblasts in cancer initiation and progression. Nature. 2004;432:332–337. doi: 10.1038/nature03096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Elenbaas B, Weinberg RA. Heterotypic signaling between epithelial tumor cells and fibroblasts in carcinoma formation. Exp. Cell Res. 2001;264:169–184. doi: 10.1006/excr.2000.5133. [DOI] [PubMed] [Google Scholar]
- 11.Brentnall TA, Lai LA, Coleman J, Bronner MP, Pan S, Chen R. Arousal of cancer associated stroma: overexpression of palladin activates fibroblasts to promote tumor invasion. PLoS ONE. 2012;7:e30219. doi: 10.1371/journal.pone.0030219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Radisky ES, Radisky DC. Matrix Metalloproteinase-Induced Epithelial-Mesenchymal Transition in Breast Cancer. J Mammary Gland Biol Neoplasia. 2010;15:201–212. doi: 10.1007/s10911-010-9177-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Christofori G. New signals from the invasive front. Nature. 2006;441:444–450. doi: 10.1038/nature04872. [DOI] [PubMed] [Google Scholar]
- 14.Nisticò P, Bissell MJ, Radisky DC. Epithelial-Mesenchymal Transition: General Principles and Pathological Relevance with Special Emphasis on the Role of Matrix Metalloproteinases. Cold Spring Harb Perspect Biol. 2012;4 doi: 10.1101/cshperspect.a011908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Desmoulière A, Geinoz A, Gabbiani F, Gabbiani G. 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;122:103–111. doi: 10.1083/jcb.122.1.103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Baum J, Duffy HS. Fibroblasts and Myofibroblasts: What are we talking about? J Cardiovasc Pharmacol. 2011;57:376–379. doi: 10.1097/FJC.0b013e3182116e39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Duffy HS. Fibroblasts, Myofibroblasts, and Fibrosis: Fact, fiction, and the Future. Journal of Cardiovascular Pharmacology. 2011;57:373–375. doi: 10.1097/FJC.0b013e3182155a38. [DOI] [PubMed] [Google Scholar]
- 18.Wipff P-J, Rifkin DB, Meister J-J, Hinz B. Myofibroblast contraction activates latent TGF-beta1 from the extracellular matrix. J. Cell Biol. 2007;179:1311–1323. doi: 10.1083/jcb.200704042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bataller R, Brenner DA. Liver fibrosis. Journal of Clinical Investigation. 2005;115:209–218. doi: 10.1172/JCI24282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gabbiani G, Hirschel BJ, Ryan GB, Statkov PR, Majno G. Granulation tissue as a contractile organ. A study of structure and function. J. Exp. Med. 1972;135:719–734. doi: 10.1084/jem.135.4.719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Churg AM, Kahn LB. Myofibroblasts and related cells in malignant fibrous and fibrohistiocytic tumors. Hum. Pathol. 1977;8:205–218. doi: 10.1016/s0046-8177(77)80081-6. [DOI] [PubMed] [Google Scholar]
- 22.Sottiurai VS, Fry WJ, Stanley JC. Ultrastructure of medial smooth muscle and myofibroblasts in human arterial dysplasia. Arch Surg. 1978;113:1280–1288. doi: 10.1001/archsurg.1978.01370230070008. [DOI] [PubMed] [Google Scholar]
- 23.Rudolph R, McClure WJ, Woodward M. Contractile fibroblasts in chronic alcoholic cirrhosis. Gastroenterology. 1979;76:704–709. [PubMed] [Google Scholar]
- 24.Gabbiani G, Majno G. Dupuytren's contracture: fibroblast contraction? An ultrastructural study. Am. J. Pathol. 1972;66:131–146. [PMC free article] [PubMed] [Google Scholar]
- 25.Martin LJ, Boyd NF. Mammographic density. Potential mechanisms of breast cancer risk associated with mammographic density: hypotheses based on epidemiological evidence. Breast Cancer Res. 2008;10:201. doi: 10.1186/bcr1831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Artinian V, Kvale PA. Cancer and interstitial lung disease. Curr Opin Pulm Med. 2004;10:425–434. doi: 10.1097/00063198-200409000-00017. [DOI] [PubMed] [Google Scholar]
- 27.Bissell DM. Chronic liver injury, TGF-beta, and cancer. Exp. Mol. Med. 2001;33:179–190. doi: 10.1038/emm.2001.31. [DOI] [PubMed] [Google Scholar]
- 28.Massagué J. TGFβ signalling in context. Nature Reviews Molecular Cell Biology. 2012;13:616–630. doi: 10.1038/nrm3434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Iyer S, Wang Z-G. Targeting TGF beta signaling for cancer therapy. Cancer Biology & Therapy. 2005;4:261–266. doi: 10.4161/cbt.4.3.1566. [DOI] [PubMed] [Google Scholar]
- 30.Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, Shappell S, Washington MK, Neilson EG, Moses HL. TGF-β Signaling in Fibroblasts Modulates the Oncogenic Potential of Adjacent Epithelia. Science. 2004;303:848–851. doi: 10.1126/science.1090922. [DOI] [PubMed] [Google Scholar]
- 31.Perera M, Tsang CS, Distel RJ, Lacy JN, Ohno-Machado L, Ricchiuti V, Samaranayake LP, Smejkal GB, Smith MG, Trachtenberg AJ, Kuo WP. TGF-β1 Interactome: Metastasis and Beyond. Cancer Genomics Proteomics. 2010;7:217–229. [PubMed] [Google Scholar]
- 32.Hinz B, Mastrangelo D, Iselin CE, Chaponnier C, Gabbiani G. Mechanical Tension Controls Granulation Tissue Contractile Activity and Myofibroblast Differentiation. Am J Pathol. 2001;159:1009–1020. doi: 10.1016/S0002-9440(10)61776-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.De Wever O, Mareel M. Role of tissue stroma in cancer cell invasion. J. Pathol. 2003;200:429–447. doi: 10.1002/path.1398. [DOI] [PubMed] [Google Scholar]
- 34.Sullivan DE, Ferris M, Nguyen H, Abboud E, Brody AR. TNF-α induces TGF-β1 expression in lung fibroblasts at the transcriptional level via AP-1 activation. Journal of Cellular and Molecular Medicine. 2009;13:1866–1876. doi: 10.1111/j.1582-4934.2008.00647.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Cat B, Stuhlmann D, Steinbrenner H, Alili L, Holtkötter O, Sies H, Brenneisen P. Enhancement of tumor invasion depends on transdifferentiation of skin fibroblasts mediated by reactive oxygen species. J Cell Sci. 2006;119:2727–2738. doi: 10.1242/jcs.03011. [DOI] [PubMed] [Google Scholar]
- 36.Wang JH-C, Thampatty BP, Lin J-S, Im H-J. Mechanoregulation of gene expression in fibroblasts. Gene. 2007;391:1–15. doi: 10.1016/j.gene.2007.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kunsch C, Medford RM. Oxidative Stress as a Regulator of Gene Expression in the Vasculature. Circulation Research. 1999;85:753–766. doi: 10.1161/01.res.85.8.753. [DOI] [PubMed] [Google Scholar]
- 38.Seoane J. Escaping from the TGFbeta anti-proliferative control. Carcinogenesis. 2006;27:2148–2156. doi: 10.1093/carcin/bgl068. [DOI] [PubMed] [Google Scholar]
- 39.Akhurst RJ, Derynck R. TGF-β signaling in cancer – a double edged sword. Trends in Cell Biology. 2001;11:S44–S51. doi: 10.1016/s0962-8924(01)02130-4. [DOI] [PubMed] [Google Scholar]
- 40.Gao D, Vahdat LT, Wong S, Chang JC, Mittal V. Microenvironmental Regulation of Epithelial–Mesenchymal Transitions in Cancer. Cancer Res. 2012;72:4883–4889. doi: 10.1158/0008-5472.CAN-12-1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hugo H, Ackland ML, Blick T, Lawrence MG, Clements JA, Williams ED, Thompson EW. Epithelial—mesenchymal and mesenchymal—epithelial transitions in carcinoma progression. Journal of Cellular Physiology. 2007;213:374–383. doi: 10.1002/jcp.21223. [DOI] [PubMed] [Google Scholar]
- 42.Giannoni E, Parri M, Chiarugi P. EMT and oxidative stress: a bidirectional interplay affecting tumor malignancy. Antioxid. Redox Signal. 2012;16:1248–1263. doi: 10.1089/ars.2011.4280. [DOI] [PubMed] [Google Scholar]
- 43.Radisky DC, Levy DD, Littlepage LE, Liu H, Nelson CM, Fata JE, Leake D, Godden EL, Albertson DG, Nieto MA, Werb Z, Bissell MJ. Rac1b and reactive oxygen species mediate MMP 3-induced EMT and genomic instability. Nature. 2005;436:123–127. doi: 10.1038/nature03688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Nieto MA. The snail superfamily of zinc-finger transcription factors. Nat Rev Mol Cell Biol. 2002;3:155–166. doi: 10.1038/nrm757. [DOI] [PubMed] [Google Scholar]
- 45.López-Novoa JM, Nieto MA. Inflammation and EMT: an alliance towards organ fibrosis and cancer progression. EMBO Mol Med. 2009;1:303–314. doi: 10.1002/emmm.200900043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R. Discovery of Endothelial to Mesenchymal Transition as a Source for Carcinoma-Associated Fibroblasts. Cancer Res. 2007;67:10123–10128. doi: 10.1158/0008-5472.CAN-07-3127. [DOI] [PubMed] [Google Scholar]
- 47.Guido C, Whitaker-Menezes D, Lin Z, Pestell RG, Howell A, Zimmers TA, Casimiro MC, Aquila S, Ando’ S, Martinez-Outschoorn UE, Sotgia F, Lisanti MP. Mitochondrial fission induces glycolytic reprogramming in cancer-associated myofibroblasts, driving stromal lactate production, and early tumor growth. Oncotarget. 2012;3:798–810. doi: 10.18632/oncotarget.574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Guido C, Whitaker-Menezes D, Capparelli C, Balliet R, Lin Z, Pestell RG, Howell A, Aquila S, Andò S, Martinez-Outschoorn U, Sotgia F, Lisanti MP. Metabolic reprogramming of cancer-associated fibroblasts by TGF-β drives tumor growth: Connecting TGF-β signaling with “Warburg-like” cancer metabolism and L-lactate production. Cell Cycle. 2012;11:3019–3035. doi: 10.4161/cc.21384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Nieman KM, Kenny HA, Penicka CV, Ladanyi A, Buell-Gutbrod R, Zillhardt MR, Romero IL, Carey MS, Mills GB, Hotamisligil GS, Yamada SD, Peter ME, Gwin K, Lengyel E. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat. Med. 2011;17:1498–1503. doi: 10.1038/nm.2492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Martinez-Outschoorn UE, Sotgia F, Lisanti MP. Power Surge: Supporting Cells “Fuel” Cancer Cell Mitochondria. Cell Metabolism. 2012;15:4–5. doi: 10.1016/j.cmet.2011.12.011. [DOI] [PubMed] [Google Scholar]
- 51.Brauer HA, Makowski L, Hoadley KA, Casbas-Hernandez P, Lang LJ, Romàn-Pèrez E, D'Arcy M, Freemerman AJ, Perou CM, Troester MA. Impact of Tumor Microenvironment and Epithelial Phenotypes on Metabolism in Breast Cancer. Clin Cancer Res. 2013;19:571–585. doi: 10.1158/1078-0432.CCR-12-2123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Rønnov-Jessen L, Petersen OW, Koteliansky VE, Bissell MJ. The origin of the myofibroblasts in breast cancer. Recapitulation of tumor environment in culture unravels diversity and implicates converted fibroblasts and recruited smooth muscle cells. J Clin Invest. 1995;95:859–873. doi: 10.1172/JCI117736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Yazhou C, Wenlv S, Weidong Z, Licun W. Clinicopathological Significance of Stromal Myofibroblasts in Invasive Ductal Carcinoma of the Breast. Tumor Biology. 2004;25:290–295. doi: 10.1159/000081394. [DOI] [PubMed] [Google Scholar]
- 54.Hennessy BT, Gonzalez-Angulo A-M, Stemke-Hale K, Gilcrease MZ, Krishnamurthy S, Lee J-S, Fridlyand J, Sahin A, Agarwal R, Joy C, Liu W, Stivers D, Baggerly K, Carey M, Lluch A, Monteagudo C, He X, Weigman V, Fan C, Palazzo J, Hortobagyi GN, Nolden LK, Wang NJ, Valero V, Gray JW, Perou CM, Mills GB. Characterization of a Naturally Occurring Breast Cancer Subset Enriched in Epithelial-to-Mesenchymal Transition and Stem Cell Characteristics. Cancer Res. 2009;69:4116–4124. doi: 10.1158/0008-5472.CAN-08-3441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Al-Ansari MM, Hendrayani SF, Shehata AI, Aboussekhra A. p16INK4A Represses the paracrine tumor-promoting effects of breast stromal fibroblasts. Oncogene. 2012 doi: 10.1038/onc.2012.270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Matise LA, Palmer TD, Ashby WJ, Nashabi A, Chytil A, Aakre M, Pickup MW, Gorska AE, Zijlstra A, Moses HL. Lack of transforming growth factor-β signaling promotes collective cancer cell invasion through tumor stromal crosstalk. Breast cancer research: BCR. 2012;14:R98. doi: 10.1186/bcr3217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kim BG, Gao M-Q, Choi YP, Kang S, Park HR, Kang KS, Cho NH. Invasive breast cancer induces laminin-332 upregulation and integrin β4 neoexpression in myofibroblasts to confer an anoikis-resistant phenotype during tissue remodeling. Breast Cancer Res. 2012;14:R88. doi: 10.1186/bcr3203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Apte M, Haber P, Darby S, Rodgers S, McCaughan G, Korsten M, Pirola R, Wilson J. Pancreatic stellate cells are activated by proinflammatory cytokines: implications for pancreatic fibrogenesis. Gut. 1999;44:534–541. doi: 10.1136/gut.44.4.534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Yen TW, Aardal NP, Bronner MP, Thorning DR, Savard CE, Lee SP, Bell RH., Jr Myofibroblasts are responsible for the desmoplastic reaction surrounding human pancreatic carcinomas. Surgery. 2002;131:129–134. doi: 10.1067/msy.2002.119192. [DOI] [PubMed] [Google Scholar]
- 60.Schäfer H, Geismann C, Heneweer C, Egberts J-H, Korniienko O, Kiefel H, Moldenhauer G, Bachem MG, Kalthoff H, Altevogt P, Sebens S. Myofibroblast-induced tumorigenicity of pancreatic ductal epithelial cells is L1CAM dependent. Carcinogenesis. 2012;33:84–93. doi: 10.1093/carcin/bgr262. [DOI] [PubMed] [Google Scholar]
- 61.Geismann C, Morscheck M, Koch D, Bergmann F, Ungefroren H, Arlt A, Tsao M-S, Bachem MG, Altevogt P, Sipos B, Fölsch UR, Schäfer H, Müerköster SS. Up-regulation of L1CAM in Pancreatic Duct Cells Is Transforming Growth Factor β1– and Slug Dependent: Role in Malignant Transformation of Pancreatic Cancer. Cancer Res. 2009;69:4517–4526. doi: 10.1158/0008-5472.CAN-08-3493. [DOI] [PubMed] [Google Scholar]
- 62.Xu Z, Vonlaufen A, Phillips PA, Fiala-Beer E, Zhang X, Yang L, Biankin AV, Goldstein D, Pirola RC, Wilson JS, Apte MV. Role of pancreatic stellate cells in pancreatic cancer metastasis. Am. J. Pathol. 2010;177:2585–2596. doi: 10.2353/ajpath.2010.090899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Shields MA, Dangi-Garimella S, Redig AJ, Munshi HG. Biochemical role of the collagen-rich tumour microenvironment in pancreatic cancer progression. Biochemical Journal. 2012;441:541–552. doi: 10.1042/BJ20111240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Zhang DY, Friedman SL. Fibrosis-dependent mechanisms of hepatocarcinogenesis. Hepatology. 2012;56:769–775. doi: 10.1002/hep.25670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Park J, Kim DS, Shim TS, Lim C-M, Koh Y, Lee SD, Kim WS, Kim WD, Lee JS, Song KS. Lung cancer in patients with idiopathic pulmonary fibrosis. Eur Respir J. 2001;17:1216–1219. doi: 10.1183/09031936.01.99055301. [DOI] [PubMed] [Google Scholar]

