Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-qxdb6 Total loading time: 0 Render date: 2024-04-29T06:20:44.163Z Has data issue: false hasContentIssue false

12 - TGF-β signaling in stem cells and tumorigenesis

from Part 2.1 - Molecular pathways underlying carcinogenesis: signal transduction

Published online by Cambridge University Press:  05 February 2015

Ying Li
Affiliation:
The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
Ruth He
Affiliation:
Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
Lopa Mishra
Affiliation:
The University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

The transformation growth factor-β (TGF-β) signaling pathway is involved in many cellular processes in both the adult organism and the developing embryo, including cell growth, cell differentiation, apoptosis, cellular homeostasis, and other cellular functions, and deregulation of the pathway can result in tumor development. TGF-β signaling maintains tissue homeostasis and prevents tumorigenesis by regulating cellular proliferation, differentiation, survival, and micro-environment. Malignant cells can overcome the tumor-suppressive effects of TGF-β, usually through two different mechanisms. First, inactivation of core components of the pathway, demonstrated as frequent mutations of core proteins, results in loss of function in TGF-β signaling in many cancers. Second, downstream alterations that disable just the tumor-suppressor arm of the pathways, allow cancer cells to utilize the remainder of the TGF-β pathway for invasion and metastasis. Insight into the powerful TGF-β pathway in the context of type and stage of cancer, micro-environment, and alteration of other signaling transduction pathways within the cancer cells is crucial to decipher the role of TGF-β as tumor suppressor or promoter, followed by the development of anti-cancer therapeutics targeting TGF-β signaling.

Molecular mechanism of TGF-β signaling: ligands, receptors, and signaling molecules, the Smads

TGF-β represents a large family of pleiotrophic growth and differentiation factors that include activin/inhibins and bone morphogenetic proteins (BMPs) (1–3). These proteins mobilize a complex signaling network to control cell fate by regulating differentiation, proliferation, motility, adhesion, and apoptosis. TGF-β is represented by three isoforms, TGF-β1, -β2, and -β3, TGF-β is secreted as an inactive latent homodimeric polypeptide that is bound to other extra-cellular proteins (4–6). The mature, bioactive ligand is produced upon proteolytic cleavage of the latent complex. Binding of the active TGF-β dimer to the type I and II receptors results in the activation of type II (TβRII), which phosphorylates and activates type I (TβRI), then propagates the signals by phosphorylating Smad transcription factors (Figure 12.1). Smad proteins together comprise a unique signaling pathway with key roles in signal transduction by TGF-β. Currently, at least eight vertebrate Smads have been identified (1,7,8). They are characterized by homologous regions at their N- and C-termini known as Mad homology, MH-1 and MH-2 domains, respectively.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 119 - 134
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Derynck, R, Zhang, YE. Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature 2003;425:577–84.CrossRef
Souchelnytskyi, S, Moustakas, A, Heldin, CH. TGF-beta signaling from a three-dimensional perspective: insight into selection of partners. Trends in Cell Biology 2002;12:304–7.CrossRef
Mishra, L, Derynck, R, Mishra, B. Transforming growth factor-beta signaling in stem cells and cancer. Science 2005;310:68–71.CrossRef
Keski-Oja, J, Koli, K, von Melchner, H. TGF-beta activation by traction? Trends in Cell Biology 2004;14:657–9.
Rifkin, DB. Latent transforming growth factor-beta (TGF-beta) binding proteins: orchestrators of TGF-beta availability. Journal of Biological Chemistry 2005;280:7409–12.CrossRef
Mishra, L, Shetty, K, Tang, Y, Stuart, A, Byers, SW. The role of TGF-beta and Wnt signaling in gastrointestinal stem cells and cancer. Oncogene 2005;24:5775–89.CrossRef
Shi, Y, Massague, J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 2003;113:685–700.CrossRef
Attisano, L, Wrana, JL. Smads as transcriptional co-modulators. Current Opinion in Cell Biology 2000;12:235–43.CrossRef
Wrana, JL, Attisano, L, Wieser, R, Ventura, F, Massague, J. Mechanism of activation of the TGF-beta receptor. Nature 1994;370:341–7.CrossRef
Wrana, JL, Tran, H, Attisano, L, et al. Two distinct transmembrane serine/threonine kinases from Drosophila melanogaster form an activin receptor complex. Molecular and Cellular Biology 1994;14:944–50.CrossRef
Groppe, J, Hinck, CS, Samavarchi-Tehrani, P, et al. Cooperative assembly of TGF-beta superfamily signaling complexes is mediated by two disparate mechanisms and distinct modes of receptor binding. Molecular Cell 2008;29:157–68.CrossRef
Liu, F, Pouponnot, C, Massague, J. Dual role of the Smad4/DPC4 tumor suppressor in TGFbeta-inducible transcriptional complexes. Genes and Development 1997;11:3157–67.CrossRef
Macias–Silva, M, Abdollah, S, Hoodless, PA, et al. MADR2 is a substrate of the TGFbeta receptor and its phosphorylation is required for nuclear accumulation and signaling. Cell 1996;87:1215–24.CrossRef
Souchelnytskyi, S, Tamaki, K, Engstrom, U, et al. Phosphorylation of Ser465 and Ser467 in the C terminus of Smad2 mediates interaction with Smad4 and is required for transforming growth factor–beta signaling. Journal of Biological Chemistry 1997;272:28 107–15.CrossRef
Chen, Y, Lebrun, JJ, Vale, W. Regulation of transforming growth factor beta- and activin-induced transcription by mammalian Mad proteins. Proceedings of the National Academy of Sciences USA 1996;93:12992–7.CrossRef
Hoodless, PA, Haerry, T, Abdollah, S, et al. MADR1, a MAD-related protein that functions in BMP2 signaling pathways. Cell 1996;85:489–500.CrossRef
Kretzschmar, M, Doody, J, Massague, J. Opposing BMP and EGF signalling pathways converge on the TGF-beta family mediator Smad1. Nature 1997;389:618–22.CrossRef
Nishihara, A, Hanai, JI, Okamoto, N, et al. Role of p300, a transcriptional coactivator, in signalling of TGF-beta. Genes and Cells 1998;3:613–23.CrossRef
Abdollah, S, Macias-Silva, M, Tsukazaki, T, et al. TbetaRI phosphorylation of Smad2 on Ser465 and Ser467 is required for Smad2-Smad4 complex formation and signaling. Journal of Biological Chemistry 1997;272:27 678–85.CrossRef
Kretzschmar, M, Liu, F, Hata, A, Doody, J, Massague, J. The TGF-beta family mediator Smad1 is phosphorylated directly and activated functionally by the BMP receptor kinase. Genes and Development 1997;11:984–995.CrossRef
Chen, HB, Shen, J, Ip, YT, Xu, L. Identification of phosphatases for Smad in the BMP/DPP pathway. Genes and Development 2006;20:648–53.CrossRef
Lin, X, Duan, X, Liang, YY, et al. PPM1A functions as a Smad phosphatase to terminate TGFbeta signaling. Cell 2006;125:915–28.CrossRef
Attisano, L, Wrana, JL. Mads and Smads in TGF beta signalling. Current Opinion in Cell Biology 1998;10:188–94.CrossRef
Heldin, CH, Miyazono, K, ten Dijke, P. TGF-beta signalling from cell membrane to nucleus through SMAD proteins. Nature 1997;390:465–71.CrossRef
Kretzschmar, M, Massague, J. SMADs: mediators and regulators of TGF-beta signaling. Current Opinion in Genetics and Development 1998;8:103–11.CrossRef
Zhang, Y, Derynck, R. Regulation of Smad signalling by protein associations and signalling crosstalk. Trends in Cell Biology 1999;9:274–9.CrossRef
Broderick, P, Carvajal-Carmona, L, Pittman, AM, et al. A genome-wide association study shows that common alleles of SMAD7 influence colorectal cancer risk. Nature Genetics 2007;39:1315–17.CrossRef
Cerutti, JM, Ebina, KN, Matsuo, SE, et al. Expression of Smad4 and Smad7 in human thyroid follicular carcinoma cell lines. Journal of Endocrinological Investigation 2003;26:516–21.
Dowdy, SC, Mariani, A, Reinholz, MM, et al. Overexpression of the TGF-beta antagonist Smad7 in endometrial cancer. Gynecological Oncology 2005;96:368–73.CrossRef
Eppert, K, Scherer, SW, Ozcelik, H, et al. MADR2 maps to 18q21 and encodes a TGFbeta-regulated MAD-related protein that is functionally mutated in colorectal carcinoma. Cell 1996;86:543–52.CrossRef
Schutte, M, Hruban, RH, Hedrick, L, et al. DPC4 gene in various tumor types. Cancer Research 1996;56:2527–30.
Kim, J, Johnson, K, Chen, HJ, Carroll, S, Laughon, A. Drosophila Mad binds to DNA and directly mediates activation of vestigial by Decapentaplegic. Nature 1997;388:304–8.CrossRef
Kretzschmar, M, Doody, J, Timokhina, I, Massague, J. A mechanism of repression of TGFbeta/Smad signaling by oncogenic Ras. Genes and Development 1999;13:804–16.CrossRef
Stroschein, SL, Wang, W, Zhou, S, Zhou, Q, Luo, K. Negative feedback regulation of TGF-beta signaling by the SnoN oncoprotein. Science 1999;286:771–4.CrossRef
Feng, XH, Zhang, Y, Wu, RY, Derynck, R. The tumor suppressor Smad4/DPC4 and transcriptional adaptor CBP/p300 are coactivators for smad3 in TGF-beta-induced transcriptional activation. Genes and Development 1998;12:2153–63.CrossRef
Zhang, Y, Musci, T, Derynck, R. The tumor suppressor Smad4/DPC 4 as a central mediator of Smad function. Current Biology 1997;7:270–6.CrossRef
Lu, T, Tian, L, Han, Y, Vogelbaum, M, Stark, GR. Dose-dependent cross-talk between the transforming growth factor-beta and interleukin-1 signaling pathways. Proceedings of the National Academy of Sciences USA 2007;104:4365–70.CrossRef
Yu, L, Hebert, MC, Zhang, YE. TGF-beta receptor-activated p38 MAP kinase mediates Smad-independent TGF-beta responses. EMBO Journal 2002;21:3749–59.CrossRef
Bakin, AV, Rinehart, C, Tomlinson, AK, Arteaga, CL. p38 mitogen-activated protein kinase is required for TGFbeta-mediated fibroblastic transdifferentiation and cell migration. Journal of Cell Science 2002;115:3193–206.
Mulder, KM, Morris, SL. Activation of p21ras by transforming growth factor beta in epithelial cells. Journal of Biological Chemistry 1992;267:5029–31.
Yan, Z, Winawer, S, Friedman, E. Two different signal transduction pathways can be activated by transforming growth factor beta 1 in epithelial cells. Journal of Biological Chemistry 1994;269:13 231–7.
Zimmerman, CM, Kariapper, MS, Mathews, LS. Smad proteins physically interact with calmodulin. Journal of Biological Chemistry 1998;273:677–80.
Shen, X, Li, J, Hu, PP, et al. The activity of guanine exchange factor NET1 is essential for transforming growth factor-beta-mediated stress fiber formation. Journal of Biological Chemistry 2001;276:15 362–8.
Bakin, AV, Tomlinson, AK, Bhowmick, NA, Moses, HL, Arteaga, CL. Phosphatidylinositol 3-kinase function is required for transforming growth factor beta-mediated epithelial to mesenchymal transition and cell migration. Journal of Biological Chemistry 2000;275:36 803–10.CrossRef
Shin, I, Bakin, AV, Rodeck, U, Brunet, A, Arteaga, CL. Transforming growth factor beta enhances epithelial cell survival via Akt-dependent regulation of FKHRL1. Molecular Biology of the Cell 2001;12:3328–39.CrossRef
Vinals, F, Pouyssegur, J. Transforming growth factor beta1 (TGF-beta1) promotes endothelial cell survival during in vitro angiogenesis via an autocrine mechanism implicating TGF-alpha signaling. Molecular and Cellular Biology 2001;21:7218–30.CrossRef
Wilkes, MC, Mitchell, H, Penheiter, SG, et al. Transforming growth factor-beta activation of phosphatidylinositol 3-kinase is independent of Smad2 and Smad3 and regulates fibroblast responses via p21-activated kinase-2. Cancer Research 2005;65:10 431–40.
Lamouille, S, Derynck, R. Cell size and invasion in TGF-beta-induced epithelial to mesenchymal transition is regulated by activation of the mTOR pathway. Journal of Cell Biology 2007;178:437–51.
Katuri, V, Tang, Y, Li, C, et al. Critical interactions between TGF-beta signaling/ELF, and E-cadherin/beta-catenin mediated tumor suppression. Oncogene 2006;25:1871–86.CrossRef
Pardali, K, Moustakas, A. Actions of TGF-beta as tumor suppressor and pro-metastatic factor in human cancer. Biochimica et Biophysica Acta 2007;1775:21–62.
Tang, Y, Katuri, V, Dillner, A, et al. Disruption of transforming growth factor-beta signaling in ELF beta-spectrin-deficient mice. Science 2003;299:574–7.CrossRef
Tang, Y, Katuri, V, Srinivasan, R, et al. Transforming growth factor-beta suppresses nonmetastatic colon cancer through Smad4 and adaptor protein ELF at an early stage of tumorigenesis. Cancer Research 2005;65:4228–37.CrossRef
Mishra, L, Katuri, V, Evans, S. The role of PRAJA and ELF in TGF-beta signaling and gastric cancer. Cancer Biology and Therapy 2005;4:694–9.CrossRef
Mishra, L, Marshall, B. Adaptor proteins and ubiquinators in TGF-beta signaling. Cytokine Growth Factor Review 2006;17:75–87.CrossRef
Monga, SP, Tang, Y, Candotti, F, et al. Expansion of hepatic and hematopoietic stem cells utilizing mouse embryonic liver explants. Cell Transplantation 2001;10:81–9.
Saha, T, Vardhini, D, Tang, Y, et al. RING finger-dependent ubiquitination by PRAJA is dependent on TGF-beta and potentially defines the functional status of the tumor suppressor ELF. Oncogene 2006;25:693–705.CrossRef
Inoue, Y, Imamura, T. Regulation of TGF-beta family signaling by E3 ubiquitin ligases. Cancer Science 2008;99:2107–12.CrossRef
Wrana, JL. Crossing Smads. Science STKE 2000;2000:RE1.
Dong, C, Li, Z, Alvarez, R, Feng, XH, Goldschmidt-Clermont, PJ. Microtubule binding to Smads may regulate TGF beta activity. Molecular Cell 2000;5:27–34.CrossRef
Tsukazaki, T, Chiang, TA, Davison, AF, Attisano, L, Wrana, JL. SARA, a FYVE domain protein that recruits Smad2 to the TGFbeta receptor. Cell 1998;95:779–91.CrossRef
Itoh, S, Thorikay, M, Kowanetz, M, et al. Elucidation of Smad requirement in transforming growth factor-beta type I receptor-induced responses. Journal of Biological Chemistry 2003;278:3751–61.CrossRef
Hocevar, BA, Smine, A, Xu, XX, Howe, PH. The adaptor molecule Disabled-2 links the transforming growth factor beta receptors to the Smad pathway. EMBO Journal 2001;20:2789–801.CrossRef
Oeda, E, Oka, Y, Miyazono, K, Kawabata, M. Interaction of Drosophila inhibitors of apoptosis with thick veins, a type I serine/threonine kinase receptor for decapentaplegic. Journal of Biological Chemistry 1998;273:9353–6.CrossRef
Chen, RH, Miettinen, PJ, Maruoka, EM, Choy, L, Derynck, R. A WD-domain protein that is associated with and phosphorylated by the type II TGF-beta receptor. Nature 1995;377:548–52.CrossRef
Chen, YG, Liu, F, Massague, J. Mechanism of TGFbeta receptor inhibition by FKBP12. EMBO Journal 1997;16:3866–76.CrossRef
Wang, T, Li, BY, Danielson, PD, et al. The immunophilin FKBP12 functions as a common inhibitor of the TGF beta family type I receptors. Cell 1996;86:435–44.CrossRef
Batut, J, Schmierer, B, Cao, J, et al. Two highly related regulatory subunits of PP2A exert opposite effects on TGF-beta/Activin/Nodal signalling. Development 2008;135:2927–37.CrossRef
Valdimarsdottir, G, Goumans, MJ, Itoh, F, et al. Smad7 and protein phosphatase 1alpha are critical determinants in the duration of TGF-beta/ALK1 signaling in endothelial cells. BMC Cell Biology 2006;7:16.CrossRef
Liu, X, Elia, AE, Law, SF, et al. A novel ability of Smad3 to regulate proteasomal degradation of a Cas family member HEF1. EMBO Journal 2000;19:6759–69.CrossRef
Bonni, S, Wang, HR, Causing, CG, et al. TGF-beta induces assembly of a Smad2-Smurf2 ubiquitin ligase complex that targets SnoN for degradation. Nature Cell Biology 2001;3:587–95.CrossRef
Stroschein, SL, Bonni, S, Wrana, JL, Luo, K. Smad3 recruits the anaphase-promoting complex for ubiquitination and degradation of SnoN. Genes and Development 2001;15:2822–36.
Lin, X, Liang, M, Feng, XH. Smurf2 is a ubiquitin E3 ligase mediating proteasome-dependent degradation of Smad2 in transforming growth factor-beta signaling. Journal of Biological Chemistry 2000;275:36 818–22.
Lo, RS, Massague, J. Ubiquitin-dependent degradation of TGF-beta-activated smad2. Nature Cell Biology 1999;1:472–8.CrossRef
Zhang, Y, Chang, C, Gehling, DJ, Hemmati-Brivanlou, A, Derynck, R. Regulation of Smad degradation and activity by Smurf2, an E3 ubiquitin ligase. Proceedings of the National Academy of Sciences USA 2001;98:974–9.CrossRef
Fukuchi, M, Imamura, T, Chiba, T, et al. Ligand-dependent degradation of Smad3 by a ubiquitin ligase complex of ROC1 and associated proteins. Molecular Biology of the Cell 2001;12:1431–43.CrossRef
Wrighton, KH, Lin, X, Feng, XH. Critical regulation of TGFbeta signaling by Hsp90. Proceedings of the National Academy of Sciences USA 2008;105:9244–9.CrossRef
de Groot, RP, Kruyt, FA, van der Saag, PT, Kruijer, W. Ectopic expression of c-jun leads to differentiation of P19 embryonal carcinoma cells. EMBO Journal 1990;9:1831–7.
Keeton, MR, Curriden, SA, van Zonneveld, AJ, Loskutoff, DJ. Identification of regulatory sequences in the type 1 plasminogen activator inhibitor gene responsive to transforming growth factor beta. Journal of Biological Chemistry 1991;266:23 048–52.
Datto, MB, Li, Y, Panus, JF, et al. Transforming growth factor beta induces the cyclin-dependent kinase inhibitor p21 through a p53-independent mechanism. Proceedings of the National Academy of Sciences USA 1995;92:5545–9.CrossRef
Li, JM, Nichols, MA, Chandrasekharan, S, Xiong, Y, Wang, XF. Transforming growth factor beta activates the promoter of cyclin-dependent kinase inhibitor p15INK4B through an Sp1 consensus site. Journal of Biological Chemistry 1995;270:26 750–3.
Li, QL, Ito, K, Sakakura, C, et al. Causal relationship between the loss of RUNX3 expression and gastric cancer. Cell 2002;109:113–24.CrossRef
Chi, XZ, Yang, JO, Lee, KY, et al. RUNX3 suppresses gastric epithelial cell growth by inducing p21(WAF1/Cip1) expression in cooperation with transforming growth factor β-activated SMAD. Molecular and Cellular Biology 2005;25:8097–107.CrossRef
Birchenall-Roberts, MC, Fu, T, Bang, OS, et al. Tuberous sclerosis complex 2 gene product interacts with human SMAD proteins. A molecular link of two tumor suppressor pathways. Journal of Biological Chemistry 2004;279:25 605–13.CrossRef
Alessi, DR, Sakamoto, K, Bayascas, JR. LKB1-dependent signaling pathways. Annual Review of Biochemistry 2006;75:137–63.CrossRef
Grady, WM, Rajput, A, Myeroff, L, et al. Mutation of the type II transforming growth factor-beta receptor is coincident with the transformation of human colon adenomas to malignant carcinomas. Cancer Research 1998;58:3101–4.
Levy, L, Hill, CS. Alterations in components of the TGF-beta superfamily signaling pathways in human cancer. Cytokine Growth Factor Review 2006;17:41–58.CrossRef
Wang, D, Kanuma, T, Mizunuma, H, et al. Analysis of specific gene mutations in the transforming growth factor-beta signal transduction pathway in human ovarian cancer. Cancer Research 2000;60:4507–12.
Goggins, M, Shekher, M, Turnacioglu, K, et al. Genetic alterations of the transforming growth factor beta receptor genes in pancreatic and biliary adenocarcinomas. Cancer Research 1998;58:5329–32.
Chen, T, Yan, W, Wells, RG, et al. Novel inactivating mutations of transforming growth factor-beta type I receptor gene in head-and-neck cancer metastases. International Journal of Cancer 2001;93:653–61.
Yakicier, MC, Irmak, MB, Romano, A, Kew, M, Ozturk, M. Smad2 and Smad4 gene mutations in hepatocellular carcinoma. Oncogene 1999;18:4879–83.CrossRef
Maliekal, TT, Antony, ML, Nair, A, Paulmurugan, R, Karunagaran, D. Loss of expression, and mutations of Smad 2 and Smad 4 in human cervical cancer. Oncogene 2003;22:4889–97.CrossRef
Sjoblom, T, Jones, S, Wood, LD, et al. The consensus coding sequences of human breast and colorectal cancers. Science 2006;314:268–74.CrossRef
Hahn, SA, Schutte, M, Hoque, AT, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science 1996;271:350–3.
Derynck, R, Akhurst, RJ, Balmain, A. TGF-beta signaling in tumor suppression and cancer progression. Nature Genetics 2001;29:117–29.CrossRef
Akiyama, Y, Iwanaga, R, Saitoh, K, et al. Transforming growth factor beta type II receptor gene mutations in adenomas from hereditary nonpolyposis colorectal cancer. Gastroenterology 1997;112:33–9.CrossRef
Grady, WM, Myeroff, LL, Swinler, SE, et al. Mutational inactivation of transforming growth factor beta receptor type II in microsatellite stable colon cancers. Cancer Research 1999;59:320–4.
Markowitz, S, Wang, J, Myeroff, L, et al. Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science 1995;268:1336–8.CrossRef
Takenoshita, S, Tani, M, Nagashima, M, et al. Mutation analysis of coding sequences of the entire transforming growth factor beta type II receptor gene in sporadic human colon cancer using genomic DNA and intron primers. Oncogene 1997;14:1255–8.CrossRef
Nagatake, M, Takagi, Y, Osada, H, et al. Somatic in vivo alterations of the DPC4 gene at 18q21 in human lung cancers. Cancer Research 1996;56:2718–20.
Thiagalingam, S, Lengauer, C, Leach, FS, et al. Evaluation of candidate tumour suppressor genes on chromosome 18 in colorectal cancers. Nature Genetics 1996;13:343–6.CrossRef
Hahn, SA, Bartsch, D, Schroers, A, et al. Mutations of the DPC4/Smad4 gene in biliary tract carcinoma. Cancer Research 1998;58:1124–6.
Howe, JR, Roth, S, Ringold, JC, et al. Mutations in the SMAD4/DPC4 gene in juvenile polyposis. Science 1998;280:1086–8.CrossRef
Pierce, DF, Gorska, AE, Chytil, A, et al. Mammary tumor suppression by transforming growth factor beta 1 transgene expression. Proceedings of the National Academy of Sciences USA 1995;92:4254–8.CrossRef
Bottinger, EP, Jakubczak, JL, Haines, DC, Bagnall, K, Wakefield, LM. Transgenic mice overexpressing a dominant-negative mutant type II transforming growth factor beta receptor show enhanced tumorigenesis in the mammary gland and lung in response to the carcinogen 7,12-dimethylbenz-[a]-anthracene. Cancer Research 1997;57:5564–70.
Forrester, E, Chytil, A, Bierie, B, et al. Effect of conditional knockout of the type II TGF-beta receptor gene in mammary epithelia on mammary gland development and polyomavirus middle T antigen induced tumor formation and metastasis. Cancer Research 2005;65:2296–302.CrossRef
Tang, B, de Castro, K, Barnes, HE, et al. Loss of responsiveness to transforming growth factor beta induces malignant transformation of nontumorigenic rat prostate epithelial cells. Cancer Research 1999;59:4834–42.
Biswas, S, Chytil, A, Washington, K, et al. Transforming growth factor beta receptor type II inactivation promotes the establishment and progression of colon cancer. Cancer Research 2004;64:4687–92.CrossRef
Barnard, JA, Beauchamp, RD, Coffey, RJ, Moses, HL. Regulation of intestinal epithelial cell growth by transforming growth factor type beta. Proceedings of the National Academy of Sciences USA 1989;86:1578–82.CrossRef
Qing, J, Liu, C, Choy, L, et al. Transforming growth factor beta/Smad3 signaling regulates IRF-7 function and transcriptional activation of the beta interferon promoter. Molecular and Cellular Biology 2004;24:1411–25.CrossRef
Weinstein, DC, Marden, J, Carnevali, F, Hemmati-Brivanlou, A. FGF-mediated mesoderm induction involves the Src-family kinase Laloo. Nature 1998;394:904–8.CrossRef
Yang, X, Li, C, Xu, X, Deng, C. The tumor suppressor SMAD4/DPC4 is essential for epiblast proliferation and mesoderm induction in mice. Proceedings of the National Academy of Sciences USA 1998;95:3667–72.CrossRef
Takaku, K, Miyoshi, H, Matsunaga, A, et al. Gastric and duodenal polyps in Smad4 (Dpc4) knockout mice. Cancer Research 1999;59:6113–17.
Tang, Y, Kitisin, K, Jogunoori, W, et al. Progenitor/stem cells give rise to liver cancer due to aberrant TGF-beta and IL-6 signaling. Proceedings of the National Academy of Sciences USA 2008;105:2445–50.CrossRef
Rubtsov, YP, Rudensky, AY. TGFbeta signalling in control of T-cell-mediated self-reactivity. Nature Reviews Immunology 2007;7:443–53.CrossRef
Engle, SJ, Ormsby, I, Pawlowski, S, et al. Elimination of colon cancer in germ-free transforming growth factor beta 1-deficient mice. Cancer Research 2002;62:6362–6.
Maggio-Price, L, Treuting, P, Zeng, W, et al. Helicobacter infection is required for inflammation and colon cancer in SMAD3-deficient mice. Cancer Research 2006;66:828–38.CrossRef
Massague, J. TGFbeta in Cancer. Cell 2008;134:215–30.CrossRef
Wu, MY, Hill, CS. Tgf-beta superfamily signaling in embryonic development and homeostasis. Developmental Cell 2009;16:329–43.CrossRef
Li, MO, Flavell, RA. TGF-beta: a master of all T cell trades. Cell 2008;134:392–404.CrossRef
Hasegawa, Y, Takanashi, S, Kanehira, Y, et al. Transforming growth factor-beta1 level correlates with angiogenesis, tumor progression, and prognosis in patients with nonsmall cell lung carcinoma. Cancer 2001;91:964–71.3.0.CO;2-O>CrossRef
Ito, N, Kawata, S, Tamura, S, et al. Positive correlation of plasma transforming growth factor-beta 1 levels with tumor vascularity in hepatocellular carcinoma. Cancer Letters 1995;89:45–8.CrossRef
Thiery, JP. Epithelial–mesenchymal transitions in development and pathologies. Current Opinion in Cell Biology 2003;15:740–6.CrossRef
Mani, SA, Guo, W, Liao, MJ, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008;133:704–15.CrossRef
Derynck, R, Akhurst, RJ. Differentiation plasticity regulated by TGF-beta family proteins in development and disease. Nature Cell Biology 2007;9:1000–4.CrossRef
Ozdamar, B, Bose, R, Barrios-Rodiles, M, et al. Regulation of the polarity protein Par6 by TGFbeta receptors controls epithelial cell plasticity. Science 2005;307:1603–9.CrossRef
Kowanetz, M, Valcourt, U, Bergstrom, R, Heldin, CH, Moustakas, A. Id2 and Id3 define the potency of cell proliferation and differentiation responses to transforming growth factor beta and bone morphogenetic protein. Molecular and Cellular Biology 2004;24:4241–54.CrossRef
Zavadil, J, Cermak, L, Soto-Nieves, N, Bottinger, EP. Integration of TGF-beta/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition. EMBO Journal 2004;23:1155–65.CrossRef
Jechlinger, M, Sommer, A, Moriggl, R, et al. Autocrine PDGFR signaling promotes mammary cancer metastasis. Journal of Clinical Investigation 2006;116:1561–70.
De Wever, O, Mareel, M. Role of tissue stroma in cancer cell invasion. Journal of Pathology 2003;200:429–47.
Allinen, M, Beroukhim, R, Cai, L, et al. Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell 2004;6:17–32.CrossRef
Ishikawa, O, LeRoy, EC, Trojanowska, M. Mitogenic effect of transforming growth factor beta 1 on human fibroblasts involves the induction of platelet-derived growth factor alpha receptors. Journal of Cell Physiology 1990;145:181–6.
Thomas, DA, Massague, J. TGF-beta directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell 2005;8:369–80.CrossRef
Friese, MA, Wischhusen, J, Wick, W, et al. RNA interference targeting transforming growth factor-beta enhances NKG2D-mediated antiglioma immune response, inhibits glioma cell migration and invasiveness, and abrogates tumorigenicity in vivo. Cancer Research 2004;64:7596–603.CrossRef
Massague, J. G1 cell-cycle control and cancer. Nature 2004;432:298–306.CrossRef
Pietenpol, JA, Holt, JT, Stein, RW, Moses, HL. Transforming growth factor beta 1 suppression of c-myc gene transcription: role in inhibition of keratinocyte proliferation. Proceedings of the National Academy of Sciences USA 1990;87:3758–62.CrossRef
Polyak, K, Kato, JY, Solomon, MJ, et al. p27Kip1, a cyclin-Cdk inhibitor, links transforming growth factor-beta and contact inhibition to cell cycle arrest. Genes and Development 1994;8:9–22.CrossRef
Feng, XH, Lin, X, Derynck, R. Smad2, Smad3 and Smad4 cooperate with Sp1 to induce p15(Ink4B) transcription in response to TGF-beta. EMBO Journal 2000;19:5178–93.CrossRef
Cordenonsi, M, Dupont, S, Maretto, S, et al. Links between tumor suppressors: p53 is required for TGF-beta gene responses by cooperating with Smads. Cell 2003;113:301–14.CrossRef
Ewen, ME, Oliver, CJ, Sluss, HK, Miller, SJ, Peeper, DS. p53-dependent repression of CDK4 translation in TGF-beta-induced G1 cell-cycle arrest. Genes and Development 1995;9:204–17.CrossRef
Iavarone, A, Massague, J. Repression of the CDK activator Cdc25A and cell-cycle arrest by cytokine TGF-beta in cells lacking the CDK inhibitor p15. Nature 1997;387:417–22.CrossRef
Kang, Y, Chen, CR, Massague, J. A self-enabling TGFbeta response coupled to stress signaling: Smad engages stress response factor ATF3 for Id1 repression in epithelial cells. Molecular Cell 2003;11:915–26.CrossRef
Siegel, PM, Shu, W, Massague, J. Mad upregulation and Id2 repression accompany transforming growth factor (TGF)-beta-mediated epithelial cell growth suppression. Journal of Biological Chemistry 2003;278:35 444–50.CrossRef
Pardali, K, Kowanetz, M, Heldin, CH, Moustakas, A. Smad pathway-specific transcriptional regulation of the cell cycle inhibitor p21(WAF1/Cip1). Journal of Cell Physiology 2005;204:260–72.
Barker, N, Clevers, H. Mining the Wnt pathway for cancer therapeutics. Nature Reviews Drug Discovery 2006;5:997–1014.CrossRef
Yue, P, Turkson, J. Targeting STAT3 in cancer: how successful are we? Expert Opinion on Investigational Drugs 2009;18:45–56.
White, LK, Wright, WE, Shay, JW. Telomerase inhibitors. Trends in Biotechnology 2001;19:114–20.CrossRef
Malumbres, M, Barbacid, M. Cell cycle, CDKs and cancer: a changing paradigm. Nature Reviews Cancer 2009;9:153–66.CrossRef
Richard, S. Finn. Preliminary results of a randomized Phase 2 study of PD 0332991, a cyclin-dependent kinase 4/6 inhibitor, in combination with letrozole for first-line treatment of patients with postmenopausal, ER-positive, HER2-negative advanced breast cancer. Journal of Clinical Oncology 2011;31.Google Scholar
Dickson, MA, Tap, WD, Keohan, ML, et al. Phase II trial of the CDK4 inhibitor PD0332991 in patients with advanced CDK4-amplified well-differentiated or dedifferentiated liposarcoma. Journal of Clinical Oncology 2013;31:2024–8.CrossRefGoogle ScholarPubMed
Geoffrey, S. A first-in-human phase I study of the CDK4/6 inhibitor, LY2835219, for patients with advanced cancer. Journal of Clinical Oncology 2013;31.Google Scholar
Sudeep, G. A phase I study of selective cyclin dependent kinase inhibitor P1446A-05 administered on an intermittent schedule in patients with advanced refractory tumors. Clinical Oncology 2012;30.
Greider, CW. Telomere length regulation. Annual Review of Biochemistry 1996;65:337–365.CrossRef
Harley, CB, Futcher, AB, Greider, CW. Telomeres shorten during ageing of human fibroblasts. Nature 1990;345:458–60.CrossRef
Vaziri, H, Schachter, F, Uchida, I, et al. Loss of telomeric DNA during aging of normal and trisomy 21 human lymphocytes. American Journal of Human Geneticst 1993;52:661–7.
Counter, CM, Avilion, AA, LeFeuvre, CE, et al. Telomere shortening associated with chromosome instability is arrested in immortal cells which express telomerase activity. EMBO Journal 1992;11:1921–9.
Lacerte, A, Korah, J, Roy, M, et al. Transforming growth factor-beta inhibits telomerase through SMAD3 and E2F transcription factors. Cell Signal 2008;20:50–9.CrossRef
Baerlocher, GM. Imetelstat rapidly induces and maintains substantial hematologic and molecular responses in patients with essential thrombocythemia (ET) who are refractory or intolerant to prior therapy: preliminary Phase II results. Blood 2012;120.
Ann, HC. The telomerase inhibitor, imetelstat, rapidly reduces myeloma cancer stem cells (CSCs) in a Phase II trial. Blood 2012;120.
Shah, S, Islam, MN, Dakshanamurthy, S, et al. The molecular basis of vitamin D receptor and beta-catenin crossregulation. Molecular Cell 2006;21:799–809.CrossRef
Larriba, MJ, Valle, N, Palmer, HG, et al. The inhibition of Wnt/beta-catenin signalling by 1alpha,25-dihydroxyvitamin D3 is abrogated by Snail1 in human colon cancer cells. Endocrine-Related Cancer 2007;14:141–51.CrossRef
Simonelli, M. Phase I study of PF-03446962 (anti-ALK-1 mAb) in hepatocellular carcinoma (HCC). Journal of Clinical Oncology 2013;31.Google Scholar
Hinz, S, Pagerols-Raluy, L, Oberg, HH, et al. Foxp3 expression in pancreatic carcinoma cells as a novel mechanism of immune evasion in cancer. Cancer Research 2007;67:8344–50.CrossRef
Oettle, H. Interm results of the phase I/II study of trabedersen (AP 12009) in patients with pancreatic carcinoma malignant melanoma, or colorectal carcinoma. Journal of Clinical Oncology 2009;27:15s.Google Scholar
Bogdahn, U. Randomized, active-controlled phase IIb study with trabedersen (AP 12009) in recurrent or refractory high-grade glioma patients: basis for phase III endpoints. Journal of Clinical Oncology, ASCO Annual Meeting Proceedings (Post-Meeting Edition) 2009;27:15S.Google Scholar
Nemunaitis, JJ, Dillman, RO, Schwarzenberger, P, et al. Phase II study of a TGF-ß2 antisense gene modified allogeneic tumor cell vaccine (Lucanix) in advanced NSCLC. Journal of Clinical Oncology; ASCO Annual Meeting Proceedings (Post-Meeting Edition). 2006;24:18S.CrossRefGoogle Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×