Dual Role of Transforming Growth Factor B in Mammary Tumorigenesis and Metastatic Progression

نویسندگان

  • Rebecca S. Muraoka-Cook
  • Nancy Dumont
  • Carlos L. Arteaga
چکیده

It is generally accepted that transforming growth factor B (TGFB) is both a tumor suppressor and tumor promoter. Whereas loss or attenuation of TGFB signal transduction is permissive for transformation, introduction of dominantnegative TGFB receptors into metastatic breast cancer cells has been shown to inhibit epithelial-to-mesenchymal transition, motility, invasiveness, survival, and metastases. In addition, there is evidence that excess production and/or activation of TGFB by cancer cells can contribute to tumor progression by paracrine mechanisms involving neoangiogenesis, production of stroma and proteases, and subversion of immune surveillance mechanisms in tumor hosts. These data provide a rationale in favor of blockade of autocrine/ paracrine TGFB signaling in human mammary tumors with therapeutic intent. Several treatment approaches are currently in early clinical development and have been the focus of our laboratory. These include (1) ligand antibodies or receptor-containing fusion proteins aimed at blocking ligand binding to cognate receptors and (2) small-molecule inhibitors of the type I TGFB receptor serine/threonine kinase. Many questions remain about the viability of anti-TGFB treatment strategies, the best molecular approach (or combinations) for inhibition of TGFB function in vivo , the biochemical surrogate markers of tumor response, the molecular profiles in tumors for selection into clinical trials, and potential toxicities, among others. INTRODUCTION The transforming growth factor h (TGFh) family comprises a superfamily of ligands that includes the TGFhs, activins, and bone morphogenetic proteins. TGFh ligands play a role in cell proliferation, functional differentiation, extracellular matrix production, cell motility, and apoptosis (1). The TGFhs are secreted as small latent complexes, which must be activated in order to enable TGFh binding to receptors. The small latent complex is composed of the active COOH-terminal TGFh dimer linked noncovalently to a dimer of the latency-associated peptide. The large latent complex is the small latent complex linked via disulfide bonds to the latent TGFh binding protein. The latent TGFh binding protein glycoproteins play a central role in the processing and localization of TGFh complexes in the extracellular matrix where the bulk of TGFhs are sequestered. Release of active TGFh from matrix-associated latent complexes may require two steps: release of the complex from the extracellular matrix by proteolysis and subsequent activation by disruption of the noncovalent association between TGFh and the latency-associated peptide. This can be achieved by chemical, enzymatic, proteolytic, and hormonal mechanisms (reviewed in ref. 2). There are three mammalian TGFh isoforms, TGFh1, TGFh2, and TGFh3 which, in general, exhibit similar function in vitro , most notably on cell growth regulation, extracellular matrix production, and immune modulation (1). However, each seems to have distinct activities in vivo as evidenced by the phenotypes of mice lacking any one of the TGFh ligands. Targeted disruption of the Tgfb1 gene leads to hematopoietic and vasculogenic defects that result in death of about half of null embryos at 10 days of gestation (3). The embryos that survive succumb to a wasting syndrome and multiorgan failure due to inflammation after weaning (4). TGFh2-null mice exhibit perinatal mortality as a result of developmental abnormalities affecting the cardiopulmonary, urogenital, visual, auditory, neural, and skeletal systems (5). Mice lacking TGFh3 die in the perinatal period and exhibit abnormal lung and palate development (6, 7). The TGFhs bind to a heteromeric complex of transmembrane serine/threonine kinases, the type I and type II receptors (ThRI and ThRII). These ligands also bind a large transmembrane proteoglycan referred as the type III TGFh receptor (also called betaglycan) whose role is to present ligand to ThRII (1). Following ligand binding to ThRII, ThRI is recruited to ligandreceptor complex. This allows for the constitutively active ThRII kinase to transphosphorylate and activate the ThRI kinase (8), which, in turn, phosphorylates the receptor-regulated Smad2 and Smad3 (Fig. 1). Smad2 and Smad3 then associate with a common mediator Smad (Smad4) and translocate to the nucleus where they regulate gene transcription (9). By contrast, the inhibitory Smad7 can interact with ThRI and prevent the phosphorylation of effector Smads (10). In addition to Smads, other signaling pathways have been implicated in TGFh actions. These include the extracellular signal-regulated kinase, c-Jun NH2-terminal kinase, p38 mitogen-activated protein kinase, phosphatidylinositol-3 kinase, and Rho GTPases (reviewed in refs. 11, 12). The roles of these non-Smad pathways in mediating the cellular effects of TGFh remain to be fully characterized. TGFB IS BOTH A TUMOR SUPPRESSOR AND A TUMOR PROMOTER TGFh was originally reported to induce anchorageindependent growth of mouse fibroblasts (13). Subsequent Presented at the Fourth International Conference on Recent Advances and Future Directions in Endocrine Manipulation of Breast Cancer, July 21-22, 2004, Cambridge, Massachusetts. Grant support: RO1 CA62212 (C.L. Arteaga), Breast Cancer Specialized Program of Research Excellence grant P50 CA98131, and Vanderbilt-Ingram Cancer Center support grant CA68485. Requests for reprints: Carlos L. Arteaga, Division of Oncology, Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 PRB, Nashville, TN 37232-6307. Phone: 615-936-3524; Fax: 615-936-1790; E-mail: [email protected]. D2005 American Association for Cancer Research. Vol. 11, 937s–943s, January 15, 2005 (Suppl.) Clinical Cancer Research 937s Research. on October 4, 2017. © 2005 American Association for Cancer clincancerres.aacrjournals.org Downloaded from studies indicated that TGFh is a potent inhibitor of epithelial cell proliferation (14). Indeed, overexpression of active TGFh under the control of tissue-specific promoters in transgenic mice delays mammary gland development (Fig. 2) and has been shown to protect from carcinogenor oncogene-induced carcinomas (15). Other data indicate that the tumor suppressor role of TGFh can be explained by its ability to inhibit cell proliferation, maintain tissue architecture (16), inhibit genomic instability (17), and induce replicative senescence and apoptosis (18). A recent report indicates that a T29 ! C polymorphism in the TGFB1 gene results in increased serum levels of TGFh1 and is associated with a reduced risk of breast cancer in postmenopausal women (19). A possible cancer-preventive effect of an excess of TGFh, as indicated by these studies, may relate to its ability to induce stem cell senescence and/or block functional differentiation in some tissues (18). Loss of TGFh-mediated growth restraint has been shown to be associated with an increased risk of transformation. For example, mice with complete or partial disruption of Tgfb1 or Smad genes are prone to the development of cancers (16, 20). Attenuation of autocrine TGFh signaling by expression of a dominant-negative ThRII results in accelerated lobulo-alveolar mammary development (21), enhanced propensity for carcinogen-induced lung, mammary, and skin tumors (22, 23), and spontaneous invasive mammary carcinomas (24). Finally, mutations in the TGFBR2 gene occur in both sporadic and inherited colon cancers with microsatellite instability (25), and restoration of ThRII by transfection reverses transformation in certain colon cancer cell lines (26). However, inactivating mutations in TGFBR2 have not been found in breast cancer (27). Although low levels of ThRII protein, as measured by immunohistochemistry, identifies a cohort of women with mammary epithelial hyperplasia at increased risk for development of breast cancer (28), there is no demonstrable difference in the proportion of invasive breast cancers that express detectable levels of ThRII protein compared with preneoplastic and preinvasive lesions (29). Interestingly, however, Gobbi et al. (29) reported that all low-grade cancers but only 31% of high-grade invasive carcinomas expressed type II receptor (Fig. 3). Although these studies support the tumor suppressive role of endogenous TGFh, it should be noted that administration of exogenous TGFh has not been shown to inhibit established neoplasms in vivo , nor has the administration of a TGFh inhibitor resulted in either spontaneous tumor development or the accelerated growth of an already established cancer. On the other hand, several reports support a causal association between an excess of endogenous or exogenous TGFh and tumor progression. Administration of recombinant TGFh1 to nude mice facilitates tumor formation by estrogendependent MCF-7 cells in the absence of estrogen supplementation. In the same report, stable transfection of a TGFh1 expression vector into MCF-7 human breast cancer cells allows them to form tumors in the absence of estrogen supplementation (30). Overexpression of activated type I TGFh receptor (31) or active TGFh1 (32) under the control of the mouse mammary tumor virus promoter were recently shown to accelerate metastases from neu-induced primary mammary tumors in transgenic mice. Furthermore, both exogenous and stably transduced TGFh1 have been shown to confer motility and invasiveness to MCF-10A nontumorigenic mammary epithelial cells transfected with HER2 (erbB2; refs. 33, 34). We recently developed a triple transgenic mouse model in which expression of active TGFh1 in mammary tissues also expressing polyomavirus middle T antigen (PyVmT) can be temporally controlled using doxycycline. In these mice, doxycyclinemediated induction of active TGFh1 for as little as 2 weeks in mice bearing late PyVmT-induced tumors markedly accelerated metastases (Fig. 4; ref. 35). There is also evidence that high production and/or activation of TGFh in tumors can enhance cancer progression by autocrine and/or paracrine mechanisms (reviewed in refs. 11, 12). Overexpression of TGFh ligands has been reported in most cancers (reviewed in ref. 36). These high TGFh levels in tumor tissues correlate with markers of a more metastatic phenotype and/or poor patient outcome, and many tumor cells exhibit increased invasiveness in response to TGFh (reviewed in ref. 37). TGFh can also induce an epithelial-to-mesenchymal transition in tumor and nontumor epithelial cells (38, 39). Reexpression of ThRII in colon cancer cells with low invasive potential restores tumor cells invasiveness (40). Forced expression of dominant-active Smad2 in squamous cancer cells also results in enhanced tumor cell motility and metastatic dissemination (41). Further underscoring the tumor-promoting role of autocrine TGFh, expression of dominant-negative ThRII in metastatic cancer cells prevents epithelial-to-mesenchymal transition and inhibits motility, tumorigenicity, and metastases (reviewed in ref. 42). These data suggest that TGFh may select for more metastatic cancers. Indeed, mice overexpressing active TGFh1 in suprabasal keratinocytes develop less benign papillomas Fig. 1 Signaling pathways induced by TGFh receptors. Phosphorylation/activation of receptor-specific Smad2 and Smad3 and the common mediator Smad4 has been shown to be associated with ligand-induced inhibition of cell proliferation. The role of non-Smad pathways such as PI3K/Akt and Ras/MAPK on TGFh actions is less clear. Role of TGFh in Breast Cancer 938s Research. on October 4, 2017. © 2005 American Association for Cancer clincancerres.aacrjournals.org Downloaded from compared with controls. However, once tumors develop, the transgenic tumors rapidly acquire a spindle cell phenotype, overexpress TGFh3, and metastasize (43). More recently, overexpression of active TGFh1 or activated ThRI in the mammary gland of transgenic mice has been shown to accelerate metastases derived from neu-induced primary mammary tumors (31, 32). Parenthetically, colon cancers with inactivating mutations of the TGFBR2 gene exhibit favorable survival compared with ThRII-positive colon cancers (44), suggesting that loss of autocrine TGFh signaling may limit systemic metastases. CLINICAL DEVELOPMENT OF TGFB INHIBITORS The improved outcome of patients bearing cancers with TGFBR2 mutations suggests an argument in favor of blockade of autocrine TGFh signaling with therapeutic intent. An additional rationale can be inferred from the paracrine effects of tumor TGFhs on angiogenesis, stroma formation and remodeling, and immunosuppression. Taken together, these observations suggest that, by blocking TGFh function, one can interrupt multiple events important for tumor maintenance. Indeed, preclinical studies have proved the principle that inhibition of TGFh affects these tumor-permissive autocrine and paracrine mechanisms (42). Several strategies to block TGFh function are being pursued. One group of strategies is aimed at blocking ligand access to TGFh receptors. Two humanized monoclonal antibodies: CAT-192, specific to TGFh1 and CAT-152, against TGFh2, are in early clinical development (45). The expression of multiple TGFh isoforms in tumors suggests that a panTGFh antibody might be more effective than isoform-specific antibodies. Two pan-TGFh monoclonal antibodies, 1D11 and 2G7, have been reported (46). The 2G7 pan-TGFh neutralizing IgG2 suppresses the establishment of MDA-231 tumors and lung metastases in athymic mice and prevents the inhibition of host natural killer cell function induced by tumor inoculation (47). The antibody had no effect against MDA-231 cells in vitro , nor did it exhibit an antitumor effect in natural killer–deficient mice, suggesting that antibodymediated TGFh blockade is effective in disrupting tumor-host immunosuppressive interactions that are essential for tumor establishment and metastatic progression. Interestingly, an antibody against the ectodomain of ThRII, which would block ligand binding, has not been reported. Another approach to prevent binding of TGFh ligands is the use of recombinant fusion proteins containing the ectodomains of ThRII and ThRIII. Soluble ThRII:Fc has shown efficacy in fibrosis and metastases models (48). In MMTV/ PyVmT transgenic mice, blockade of TGFh with soluble ThRII:Fc increases apoptosis in primary mammary tumors and inhibits tumor cell motility, intravasation, and metastases (49). In this report, treatment with soluble ThRII:Fc inhibited Akt activity in tumors. Human recombinant ThRIII has shown antimetastatic and antiendothelial cell activity (50). One attractive feature of recombinant betaglycan over soluble ThRII RII is its greater affinity for TGFh2. A second group of strategies is aimed at directly blocking the receptors’ catalytic activity. SBI-14352, NPC 30345, and LY364947 (51–53) are ATP competitive inhibitors of the ATP binding site of the ThRI kinase. This approach spares the ThRII kinase and, therefore, may not inhibit TGFh function completely. If complete inhibition of TGFh was required for antitumor action, this selectivity could compromise anticancer activity but at the same time ameliorate potential toxicities. These two possibilities are theoretical, because there are no known ThRII functions that do not require ThRI. Nonetheless, the development of bifunctional ThR kinase inhibitors should help in resolving these questions. Vectors encoding the Fig. 2 Induced expression of active TGFh1 delays mammary gland morphogenesis. A constitutively active simian TGFh1 transgene under the control of the tet-op7 promoter was constructed. The tet-op7 promoter contains seven tandem repeats of a sequence that is transactivated by the transcription factor reverse-tetracycline-transactivator (rtTA ) only in the presence of doxycycline (DOX ; ref. 59). Mice expressing this transgene in their germ line were crossbred with MMTV/rtTA mice to generate double transgenics. Shown are hematoxylinstained whole mounts of #4 mammary glands from 12-week-old mice that have been kept on DOX since birth. Consistent with the tumorsuppressive effect of TGFh, glands from DOX-treated bigenic mice exhibited a severe delay in mammary ductal progression. Modified from Muraoka et al. (35). Clinical Cancer Research 939s Research. on October 4, 2017. © 2005 American Association for Cancer clincancerres.aacrjournals.org Downloaded from inhibitory Smad (Smad7) have been used to bind ThRI and interfere with Smad2 and Smad3 phosphorylation (54). This strategy should be viewed with caution in that it will not block Smad-independent TGFh-induced responses conducive to tumor progression. Indeed, Smad7 mRNA is overexpressed in pancreatic cancers and its forced expression in pancreatic cancer cells results in loss of TGFh-mediated growth inhibition but facilitates anchorage-independent growth and tumorigenicity (55). Moreover, blockade of Smad4 has been shown to facilitate TGFh-dependent and TGFh-independent activation of Erk in squamous cancer cells and promote their motility and transmesenchymal differentiation (56). The TGFh antagonists discussed above are summarized in Fig. 5. A potential risk of therapy with TGFh antagonists is the acceleration of preneoplastic lesions or cancers in which TGFh still exerts growth restraint. In addition to having an antitumor effect, blockade of TGFh in normal cells may induce side effects in the tumor host. Complete loss of TGFh in mice is associated with a severe inflammatory response (4). Complete elimination of TGFh function in T cells leads to autoimmune disorders (57). A recent experiment, however, suggests that TGFh antagonists might be well tolerated. Mice expressing soluble ThRII under the regulation of the mouse mammary tumor virus/long terminal repeat promoter exhibit high levels of the TGFh antagonist in the circulation without the severe inflammatory phenotype of TGFh-null mice. Interestingly, the circulating levels of sThRII:Fc were enough to inhibit tumor metastases in this model. Mild lymphocytic infiltration in lungs, kidneys, and pancreas were observed but no spontaneous tumors developed (58). It is likely that the severe inflammatory and autoimmune phenotype observed in genetically engineered mice reflects the complete loss of TGFh function, a level of inhibition that is unlikely to be achieved with exogenous inhibitors. Fig. 3 A, type II TGFh receptor expression in preneoplastic lesions and invasive breast cancers. ThRII was detected by immunohistochemistry in archival breast specimens including benign proliferative lesions, ductal carcinoma in situ (DCIS), and invasive mammary carcinomas (n = 187). Although cancer specimens showed reduced expression of ThRII in neoplastic cells compared with normal mammary cells and benign lesions, this reduction did not achieve statistical significance. B, ThRII protein expression and histological grade. There was a significant inverse correlation between undetectability of ThRII protein and tumor grade (P = 0.001) in invasive breast cancers (n = 43). Modified from Gobbi et al. (29). Fig. 4 Induction of TGFh1 increases lung metastases in transgenic mice. Mice expressing middle T antigen in the mammary gland (MMTV/PyVmT) were cross-bred with MMTV/rtTA tet-op/TGFh1 bigenics. Triple transgenic mice were treated with DOX in the drinking water starting at week 9. The majority of DOX-treated mice exhibited signs of respiratory distress at 13 weeks. Shown are low-power H&E sections of lungs from triple transgenic mice treated or not with DOX from weeks 9 to 13. DOX-treated mice displayed an average of 162 F 15.9 (n = 15) surface lung metastases compared with 17.6 F 2.6 (n = 11) in untreated controls. Role of TGFh in Breast Cancer 940s Research. on October 4, 2017. © 2005 American Association for Cancer clincancerres.aacrjournals.org Downloaded from CONCLUSIONS Significant experimental evidence suggests that TGFh can foster tumor-host interactions that indirectly support the viability and progression of neoplastic cells. Furthermore, autocrine TGFh signaling is operative in some tumor cells and can contribute to tumor invasion, survival, and metastases. This possibility is likely in a cohort of women with breast cancers in which loss of TGFh receptors and/or signal transducers is uncommon. The multiple tumor-permissive effects of TGFh provide a therapeutic opportunity, in that blocking this signaling network interrupts several autocrine and paracrine mechanisms that are essential for tumor maintenance. OPEN DISCUSSION Dr. Kent Osborne: One of the things TGFh does is activate MMPs. So one of the mechanisms, then, of activation of MAP kinase and Akt may be by activation of MMP2, cleavage of heparin-binding EGF and autocrine activation of EGF receptor. Have you tried blocking the EGF receptor and seeing if you can activate that? Dr. Carlos Arteaga: Yes, we have. Blockade of the EGF receptor tyrosine kinase with gefitinib blocks TGFh-induced motility completely. In A431 and NMuMG cells, TGFh can activate MMP2 and MMP9 while the levels of secreted TGFa and amphiregulin go way up. We can block this response by using inhibitors of TACE, TNFa-converting enzyme, which cleaves membrane-tethered TGFa and amphiregulin. So, TGFh receptors are functioning just like G-protein coupled receptors, in that they activate TACE, which cleaves membrane-tethered ligands, which then activate the EGFR. Dr. Osborne: It’s like the story with estrogen treatment of those cells in the setting of high HER2 or high EGFR: you get Src activation, MMP2 activation, and the same kind of autocrine-mediated effects. Dr. Richard Santen: The EGF receptor when activated can bind to focal adhesion kinase, which then turns on downstream PAK and Rac, which are involved in invasiveness. In follow-up to Dr. Osborne’s question, if the heparin-binding EGF is freed up and binds to the EGF receptor, under those circumstances can you demonstrate either EGF receptor binding to focal adhesion kinase or the tyrosine 397 phosphorylation on focal adhesion kinase? Have you looked at that? Dr. Arteaga: We have not looked specifically at what you are asking but we have a recent paper that addresses a related issue [J Biol Chem 2004;279:24505–13]. In HER2transfected MCF-10A mammary epithelial cells, but not in control cells with low levels of HER2, TGFh induces motility and invasiveness. In the control cells it just inhibits their proliferation. So here we have a proto-oncogene, HER2, unmasking the ability of TGFh to induce motility and potentially accelerate metastatic progression. In the transfected cells, the Rac1 GTPase is very potently activated within 15 minutes but, interestingly, Rac1 is also activated in the control cells. What is different is that the control cells do not move, whereas the HER2-overexpressing cells do. In addition, we see an association between Rac1, PAK, HER2, actin, actinin, and the type II TGFh receptor at the leading edges of TGFhstimulated cells. However, we have not looked at FAK, at focal adhesions, yet as you suggest. Dr. Santen: How do you explain two cells that have increased Rac, one of which is motile whereas the other isn’t? Dr. Arteaga: We speculate that, in addition to Rac, there are other cellular determinants activated by HER2 signaling in the proto-oncogene overexpressing cells that are required for cell motility. The recruitment of additional transducers to a GTPaseHER2 complex may be very important for transformed cell motility. Dr. Jose Russo: Could it have been that the observations in transgenic mice might be an artifact and is not a reflection of the human situation? It could be that we are developing a biology and understanding of TGFh exclusively for the mouse that is not applicable to humans. Dr. Arteaga: I presume you are skeptical of the increasing number of studies in transgenic mice supporting a role for TGFh in metastatic progression. Your presumption is certainly plausible. However, there is a recent interesting report by Knabbe et al. [Clin Cancer Res 2004;10:491–8] that shows that ER-negative tumors that lack the type II TGFh receptor have a good prognosis, similar to ER+ tumors, further supporting the possibility of an association between autocrine TGFh signaling and a more metastatic phenotype. Then there are the data with microsatellite unstable colon cancers that go along the same lines. In my opinion, the soon-to-be-initiated therapeutic trials with TGFh inhibitors will determine in the end if some tumors are driven by autocrine/paracrine TGFh, as the preclinical data suggest. Dr. Douglas Yee: You mentioned a study reporting a polymorphism that reduces the amount of h1. So it’s detectable in the serum? How much circulating TGFh is there? Dr. Arteaga: Yes, it is detectable in serum. The paper by Ziv et al. in JAMA [2001;285:2859–63] indicates a polymorphism associated with increased levels of serum TGFh and a markedly reduced risk of breast cancer. It is difficult to know if that circulating pool reflects what is happening in tumor and nontumor tissues. TGFhs are ubiquitous and made as latent proteins. These latent ligands accumulate in very high levels in the extracellular space where Fig. 5 Sites of action of TGFh antagonists currently in development. Smad7 is not included. There are no reports of TGFh receptor antibodies that block ligand binding in clinical development. Because of the low affinity of the type II receptor for TGFh2, the soluble ThRII:Fc fusion protein should inhibit TGFh1 and TGFh3 but not TGFh2. Clinical inhibitors ofxTGFh activation have likewise not been reported. Clinical Cancer Research 941s Research. on October 4, 2017. © 2005 American Association for Cancer clincancerres.aacrjournals.org Downloaded from they can be activated in a temporally and spatially regulated manner. So, how the circulating levels reflect local activation and function is unclear. I should add that we don’t know much about the physiologic and pathologic determinants of TGFh activation in situ . Dr. Yee: So no one has attempted to look at TGFh levels and cancer risk in the big serum banks like the Nurses’ Health Study, for example? Dr. Arteaga: To my knowledge, nobody has looked at this specific cohort, perhaps in part because of the limitations of the methods and the difficulty in the interpretation of any results. Another issue is that TGFh activation could have occurred in vitro but not in the patient. On the other hand, if you measure TGFh ligands, how do you know they are active in situ? We don’t have good reagents that will look specifically at ‘‘active’’ TGFh or activation of TGFh signal transduction in vivo and, therefore, offer some assurances that the serum level correlates with TGFh functions at tissue sites.

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

ثبت نام

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

منابع مشابه

Tumor Formation and Metastasis Development and Polyomavirus Middle T Antigen Induced Gene in Mammary Epithelia on Mammary Gland

Transforming growth factor–B (TGF-B) isoforms are growth factors that function physiologically to regulate development, cellular proliferation, and immune responses. The role of TGF-B signaling in mammary tumorigenesis is complex, as TGF-B has been reported to function as both a tumor suppressor and tumor promoter. To elucidate the role of TGF-B signaling in mammary gland development, tumorigen...

متن کامل

Enhanced Hepatocyte Growth Factor Signaling by Type II

Transforming growth factor-B (TGF-B) plays complex dual roles as an inhibitor and promoter of tumor progression. Although the influence of the stromal microenvironment on tumor progression is well recognized, little is known about the functions of TGF-B signaling in the stroma during tumor progression. Using cre-lox technology, expression of the type II TGF-B receptor was selectively knocked ou...

متن کامل

Enhanced hepatocyte growth factor signaling by type II transforming growth factor-beta receptor knockout fibroblasts promotes mammary tumorigenesis.

Transforming growth factor-beta (TGF-beta) plays complex dual roles as an inhibitor and promoter of tumor progression. Although the influence of the stromal microenvironment on tumor progression is well recognized, little is known about the functions of TGF-beta signaling in the stroma during tumor progression. Using cre-lox technology, expression of the type II TGF-beta receptor was selectivel...

متن کامل

Dual role of transforming growth factor beta in mammary tumorigenesis and metastatic progression.

It is generally accepted that transforming growth factor beta (TGFbeta) is both a tumor suppressor and tumor promoter. Whereas loss or attenuation of TGFbeta signal transduction is permissive for transformation, introduction of dominant-negative TGFbeta receptors into metastatic breast cancer cells has been shown to inhibit epithelial-to-mesenchymal transition, motility, invasiveness, survival,...

متن کامل

Effect of Transforming Growth Factor-β (TGF-β) on proliferation of gastric epithelial cells in culture

Objective: Helicobacter pylori has a well-established role in the development of gastric cancer. In vitro studies reveal increased proliferation of the gastric mucosa in the presence of H. pylori infection. It has been also shown that production of some cytokines, such as interleukin-1 beta (IL-1b) is in...

متن کامل

TGFβ: A player on multiple fronts in the tumor microenvironment.

The physiological functions of transforming growth factor (TGF)-β in cell signaling include regulation of developmental processes and cell growth. Tumor cells very often display altered regulation of the TGFβ signaling pathway, either by defects in TGFβ itself or in downstream components of the pathway. TGFβ can play a dual role in tumorigenesis, i.e. it can be either tumor-suppressive or tumor...

متن کامل

ذخیره در منابع من


  با ذخیره ی این منبع در منابع من، دسترسی به آن را برای استفاده های بعدی آسان تر کنید

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

ثبت نام

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

عنوان ژورنال:

دوره   شماره 

صفحات  -

تاریخ انتشار 2005