The TGF-β Paradox in Human Cancer: An Update (original) (raw)

Future Oncol. Author manuscript; available in PMC 2009 Jul 15.

Published in final edited form as:

PMCID: PMC2710615

NIHMSID: NIHMS113160

Author Affiliation: Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045

Correspondence: William P. Schiemann, Department of Pharmacology, MS-8303, University of Colorado Denver, Anschutz Medical Campus, RC1 South Tower, Room L18-6110, 12801 East 17th Avenue, PO Box 6511, Aurora, CO 80045. Phone: (303)724-1541. Fax: (303)724-3663. E-mail: ude.cshcu@nnameihcS.lliB

Summary

Transforming growth factor-β (TGF-β) plays an essential role in maintaining tissue homeostasis through its ability to induce cell cycle arrest, differentiation, apoptosis, and to preserve genomic stability. Thus, TGF-β is a potent anticancer agent that prohibits the uncontrolled proliferation of epithelial, endothelial, and hematopoietic cells. Interestingly, tumorigenesis typically elicits aberrations in the TGF-β signaling pathway that engenders resistance to the cytostatic activities of TGF-β, thereby enhancing the development and progression of human malignances. Moreover, these genetic and epigenetic events conspire to convert TGF-β from a suppressor of tumor formation to a promoter of their growth, invasion, and metastasis. The dichotomous nature of TGF-β during tumorigenesis is known as the “TGF-β Paradox,” which remains the most critical and mysterious question concerning the physiopathological role of this multifunctional cytokine. Here we review recent findings that directly impact our understanding of the “TGF-β Paradox” and discuss their importance to targeting the oncogenic activities of TGF-β in developing and progressing neoplasms.

Keywords: Angiogenesis, Cancer, Cell Invasion, Epithelial-mesenchymal Transition, Metastasis, Signal Transduction, Transforming growth factor-β

1. TGF-β and the Tumor Microenvironment

1.1. TGF-β and Fibroblasts

Tumor development in many respects mirrors that of an organ, albeit in a highly dysfunctional and disorganized manner. For instance, whereas normal tissue specification requires reciprocal signaling inputs from distinct cell types and matrix proteins, the phenotype of developing carcinomas is similarly dictated by the dynamic interplay between malignant cells and their accompanying stroma, which houses fibroblasts and endothelial cells (ECs), as well as a variety of infiltrating immune and progenitor cells [1, 2]. Moreover, tumor reactive stroma not only plays an important role during cancer initiation and progression, but also in determining whether TGF-β suppresses or promotes tumor formation (Figure 1; [35]). Along these lines, TGF-β exerts its anti-tumor activities by regulating epithelial cell behavior, and by regulating that of adjacent fibroblasts, which synthesize and secrete a variety of cytokines, growth factors, and extracellular matrix (ECM) proteins that mediate tissue homeostasis and suppress cancer development. Thus, inactivating paracrine TGF-β signaling between adjacent epithelial and stromal compartments promotes cellular transformation, as well as induces the growth, survival, and motility of developing neoplasms [6, 7]. For instance, rendering fibroblasts deficient in the expression of the TGF-β type II receptor (TβR-II), which manifests as insensitivity to TGF-β, results in the formation of prostate intraepithelial neoplasia and invasive carcinoma of the forestomach [3]. Conditional deletion of TβR-II in mammary gland fibroblasts enhanced their proliferation and abundance within abnormally developed ductal units [8]. Interestingly, grafting a mixture of TβR-II-deficient mammary fibroblasts with mammary carcinoma cells under the subrenal capsule significantly enhanced the growth and invasion of breast cancer cells. The enhanced tumorigenicity of implanted mammary carcinoma cells was not recapitulated in grafts containing TGF-β-responsive fibroblasts, which failed to synthesize and secrete the high levels of TGF-α, MSP (macrophage-stimulating protein), and HGF (hepatocyte growth factor) produced by their TβR-II-deficient counterparts [35, 8]. Thus, TGF-β signaling in fibroblasts suppresses their activation of cancer-promoting paracrine signaling axes that target adjacent epithelial cells. Somewhat surprisingly, TβR-II-deletion in mammary carcinoma cells resulted in the activation of two tumorigenic paracrine signaling axes comprised of SDF-1:CXCR4 and CXCL5:CXCR2, which collectively function in recruiting immature GR1+CD11b+ myeloid cells to developing mammary tumors [9]. Upon their arrival within mammary tumor microenvironments, GR1+CD11b+ cells promote breast cancer cell invasion and metastasis by attenuating host tumor immunosurveillance, and by stimulating MMP expression [9]. Recently, the ability of TGF-β to induce cell cycle progression in glioma cells required initiation of autocrine PDGF-B signaling. Importantly, the proliferation promoting properties of TGF-β and Smad2/3 only occurred in glioma lacking methylation of the PDGF-B gene, suggesting that the methylation status of PDGF-B determines the oncogenic activities of TGF-β in part via autocrine PDGF-B signaling within tumor microenvironments [10].

An external file that holds a picture, illustration, etc. Object name is nihms113160f1.jpg

Cellular Targets of TGF-β During the Development and Progression of Human Cancers

TGF-β is a multifunctional cytokine that normally suppresses cell proliferation, differentiation, and apoptosis, as well as regulates cell and tissue homeostasis. Under normal physiological conditions, TGF-β functions as a tumor suppressor by preventing the ability of cells to progress through the cell cycle, or by stimulating the ability of cells to undergo apoptosis or differentiation. However, genetic and epigenetic events that transpire during tumorigenesis can convert TGF-β from a tumor suppressor to a tumor promoter, particularly the ability of cancer cells to acquire invasive and metastatic phenotypes. The oncogenic activities of TGF-β also are coordinated by dysregulated autocrine and paracrine signaling networks that take place between epithelial, fibroblasts, endothelial, and immune cells, that collectively promote tumor angiogenesis, invasion, and metastasis, and inhibit host immunosurveillance within tumor microenvironments. See text for specific examples of how TGF-β signaling becomes dysregulated during tumorigenesis

Tumorigenesis often is accompanied by intense desmoplastic and fibrotic reactions, which elicit the formation of rigid tumor microenvironments that enhance the selection and expansion of metastatic cells [11, 12]. Lysyl oxidases (LOX) belong to a 5 gene family of copper-dependent amine oxidases (i.e., LOX, LOXL, LOXL2, LOXL3, and LOXL4) that function in cross-linking collagens to elastin in the ECM [13, 14]. Mechanistically, the activation of these cross-linking reactions by LOXs secreted by fibroblasts and epithelial cells serves to increase the tensile strength and structural integrity of tissues during embryonic development and organogenesis, as well as during the maintenance of normal tissue homeostasis [13, 14]. Similar to TGF-β, members of the LOX family have been associated with tumor suppression and tumor promotion. Indeed, the transformation of fibroblasts by oncogenic Ras is suppressed by LOX and its ability to bind, oxidize, and inactivate growth factors housed in cell microenvironments, which presumably contributes the loss of cyclin D1 expression observed in LOX-expressing fibroblasts [15, 16]. More recently, LOX was observed to interact physically with TGF-β1 and alter its ability to stimulate Smad3 in cultured osteoblasts [17], while LOXL4 expression inhibited TGF-β stimulation of liver cancer cell invasion through synthetic basement membranes [18]. Thus, these findings implicate LOXs as potential suppressive agents within tumor microenvironments. In stark contrast, aberrant LOX activity also is associated with cancer progression, particularly the selection, expansion, and dissemination of metastatic cells [1315, 1922]. Indeed, upregulated LOX expression is (i) essential for hypoxia-induced metastasis of human MDA-MB-231 breast cancer cells in mice [19]; and (ii) is observed most frequently in poorly differentiated, high grade mammary tumors and, consequently, predicts for increased disease recurrence and decreased patient survival [15, 19]. Recently, we observed LOX expression to be induced strongly by TGF-β in normal and malignant MECs, and in mammary tumors produced in mice. Moreover, inhibiting LOX activity or degrading its metabolic byproduct, hydrogen peroxide, both antagonize the ability of TGF-β to induce the proliferation, EMT, and invasion in normal and malignant MECs. Furthermore, we find that LOX antagonism uncouples TGF-β from stimulating Src and p38 MAPK (M. Taylor and W.P. Schiemann, unpublished findings), whose activities are essential for mediating oncogenic signaling by TGF-β in breast cancer cells [2325]. Along these lines, future studies need to enhance our understanding of (i) the role of tumor reactive fibroblasts and their production of TGF-β in protecting carcinoma cells from tumoricidal radiotherapies, and (ii) the molecular and cellular mechanisms whereby anti-TGF-β therapies selectively sensitize carcinoma cells, not their adjacent normal counterparts, to ionizing radiation treatments [2628].

Collectively, these findings highlight the important role TGF-β plays in governing autocrine and paracrine signaling networks, and more importantly, demonstrate how disrupting the delicate balance between these systems contributes to carcinoma development and progression.

1.2. TGF-β and Immunosurveillance

In addition to its regulation of stromal fibroblasts, TGF-β present in cell microenvironments also plays an essential role in governing the delicate balance between host immunosurveillance and inflammation, which collectively can determine whether tumor development and progression is induced or inhibited [29, 30]. The importance of TGF-β in regulating immune system function and homeostasis is underscored by the finding that (i) TGF-β1-deficient mice exhibit lethal multifocal inflammatory disease [31, 32]; (ii) Smad3-deficient mice exhibit defects in the responsiveness and chemotaxis of their neutrophils, and their T and B cells [33]; and (iii) transgenic expression of truncated TβR-II specifically in T cells results in severe autoimmune reactions characterized by multifocal inflammation and autoantibody production [31]. Furthermore, T cell-specific deletion of Smad4 in mice drives T cell differentiation towards a Th2 phenotype and their elevated secretion of interleukins (ILs) 4, 5, 6, and 13 [34]. Similar to fibroblasts, the net effect of disrupting paracrine T cell signaling networks is the development of gastrointestinal carcinomas in these genetically engineered animals [34]. In addition, cancer cells typically increase their production and secretion of TGF-β into tumor microenvironments, as well as into the general circulation of cancer patients [3537]. Abnormally elevated TGF-β concentrations also are detected within the tumor milieu in response to ECM degradation mediated by resident and recruited leukocytes – i.e., monocytes/macrophages, dendritic cells, granulocytes, mast cells, T cells, and natural killer (NK) cells – that either promote or suppress tumor development in a context-specific manner [38].

1.2.1. TGF-β and Adaptive Immunity

TGF-β suppresses host immunosurveillance by inhibiting the proliferation and differentiation of NK and T cells, and by diminishing their synthesis and secretion of cytotoxic effector molecules, including interferon-γ, lymphotoxin-α, perforin/granzyme, and Fas ligand [30, 39, 40]. TGF-β also inhibits the tumor-targeting activities of T and NK cells through its stimulation of Tregs housed within tumor microenvironments [41]. Whereas TGF-β potently inhibits the proliferation of naïve CD8+ T cells, this cytokine elicits little-to-no activity in fully differentiated CD8+ T cells due to their downregulation of TβR-II. Administration of ILs 2 or 10 to differentiated CD8+ T cells restores their responsiveness to TGF-β, as does expression of the co-stimulatory molecule CD28, which promotes the survival of memory/effector phenotypes in thymic and peripheral T cell populations [30, 39, 42]. Mechanistically, the immunosuppressive effects of TGF-β transpire in part via Smad3, whose phosphorylation and activation prevents the mitogenesis of CD8+ T cells by (i) inhibiting their production of IL-2; (ii) repressing their expression of c-Myc, cyclin D2, and cyclin E; and (iii) stimulating the expression of the CDKIs p15, p21, and p27 [30, 39, 40]. In contrast to its stimulation of cytostasis in CD8+ T cells, TGF-β has no effect on the proliferation of CD4+ T cells, but does inhibit the differentiation of CD4+ T cells into Th1 and Th2 lineages by (i) downregulating T cell receptor expression; (ii) reducing intracellular Ca++ signaling; and (iii) repressing the expression and activation of transcription factors [30, 39, 40], all of which weaken host immunosurveillance. Collectively, these findings predict that inactivating TGF-β signaling in CD8+ or CD4+ T cells will inhibit tumor formation by elevating host immunosurveillance, a supposition shown to occur during T cell-mediated eradication of skin [43] and prostate [44] cancers in mice. More recently, TGF-β was observed to promote the development and progression of breast and colon cancers by inducing CD8+ T cells to secrete IL-17, which exerts pro-survival signaling in carcinoma cells [45]. Thus, in addition to improving host immunosurveillance, neutralizing TGF-β function in T cells also will improve tumor resolution by suppressing the activation of carcinoma survival pathways.

1.2.2. TGF-β and Innate Immunity

In addition to its role in regulating adaptive immunity, TGF-β also plays an essential role in directing activities and behaviors of components of the innate immune system, including NK cells, dendritic cells, mast cells, monocytes, and macrophages. Indeed, we defined a novel TAB1:xIAP:TAK1:IKKβ:NF-κB signaling axis that forms aberrantly in breast cancer cells, and in normal MECs following their induction of EMT by TGF-β. Once formed, this signaling axis enables oncogenic signaling by TGF-β in part via activation of NF-κB and its consequential production of proinflammatory cytokines, which promote breast cancer growth in mice in a manner consistent with regulation of innate immunity by TGF-β [46]. Along these lines, TGF-β receptors were observed to associate with those for IL-1β, thereby enabling (i) TGF-β to activate NF-κB; (ii) IL-1β to activate Smad2; and (iii) both pathways to potentiate inflammatory cytokine production [47] their ability to promote inflammation and the enhanced the survival of tumor-associated monocytes [48, 49]. In addition, transgenic expression of IL-1β in the stomachs of mice promoted the activation of myeloid-derived suppressor cells (MDSCs) via an IL-1R/NF-κB signaling axis, whose inappropriate and constitutive activation results in the formation of stomach neoplasias [50]. TGF-β is a potent inhibitor of the cytolytic activity of NK cells, presumably by attenuating the activation of their NKp30 and NKD2D receptors, and by inhibiting their production of interferon-γ. In addition, TGF-β also represses the activities of dendritic cells by inhibiting their expression of MHC class II, CD40, CD80, and CD86, and TNF-α, IL-12, and CCL5/Rantes [30, 39, 40, 51]. Mast cells are actively recruited to tumor microenvironments by TGF-β where they synthesize and secrete numerous tumor promoting factors, including histamine, proteases, and cytokines (e.g., VEGF and TGF-β) [40, 52]. Lastly, TGF-β stimulates monocytes and macrophage chemotaxis to tumor microenvironments, leading to enhanced tumor invasion, angiogenesis, and metastasis, and to diminished antigen presentation and immunosurveillance towards developing neoplasms [53, 54].

1.3. TGF-β and Endothelial Cells

Angiogenesis is the process whereby new blood vessels sprout and form from preexisting vessels; it also is an essential physiological process that transpires during embryonic development, wound healing, and the female reproductive cycle [55, 56]. The initiation of pathological angiogenesis has been linked to numerous human diseases, including rheumatoid arthritis, diabetic retinopathy, and age-related macular degeneration [56, 57]. Interestingly, all solid tumors larger than 1 cm3 suffer from hypoxia [58], and as such, initiate angiogenesis as a means of acquiring an efficient supply of nutrients and waste removal, as well as a route for their metastasis to distant locales. Two distinct phases are involved in angiogenesis, namely angiogenic activation and resolution. During the activation phase of angiogenesis, ECs initially exhibit increased vessel permeability and elevated rates of cell proliferation, migration, and invasion. In addition, new vessel sprouting is further enhanced by a reduction in EC adhesion, coupled to an alteration in basement membrane integrity. In contrast, angiogenic resolution essentially restores activated ECs to their resting, quiescent phenotypes, as well as promotes the recruitment of perivascular cells that maintain vessel stability and hemodynamics [5557].

TGF-β plays critical roles in regulating both the activation and resolution phases of angiogenesis [5962]. Indeed, homozygous deletion of various components of the TGF-β signaling system in mice routinely results in the appearance of vascular and EC defects, particularly in animals lacking TGF-β1 [63], TβR-I [64], TβR-II [65, 66], TβR-III [67, 68], Smad1 [69], or Smad5 [70]. In humans, loss or inactivation of endoglin leads to hereditary hemorrhagic telangiectasia type 1 (HHT1) [71, 72], while that of ALK1 results in HHT2 [73, 74]. Moreover, the defects associated with HHT1 and HHT2 in humans are phenocopied in knockout mice lacking expression of either endoglin [75, 76] or ALK1 [7779], respectively. Thus, altered expression and/or activity of TGF-β in tumor microenvironments clearly will impact the ability of hypoxic tumors overcome this impediment to their growth and survival.

ECs have been reported to express two distinct TβR-Is, namely TβR-I/Alk5 and ALK1. The importance of these two receptors in mediating vessel development by TGF-β is evidenced by the embryonic lethality observed at day E11.5 and E10.5 in mice lacking ALK1 [79] or ALK5 [64], respectively. Recent evidence also suggests that these two type I receptors differentially regulate the coupling of TGF-β to angiogenic activation and resolution. For instance, TβRI/ALK5 activation stimulates Smad2/3 and the subsequent expression of genes operant in mediating vessel maturation, including plasminogen activator inhibitor 1 (PAI-1) and fibronectin [78, 80, 81]. Moreover, microarray gene expression analyses of EC cells before and after their stimulation with TGF-β confirmed that the activation of a TGF-β:TβR-I/ALK5:Smad2/3 signaling axis does indeed promote angiogenic resolution [61, 82]. In contrast, ALK1 activation stimulates Smad1/5 and the subsequent expression of genes operant in mediating angiogenesis activation, including Id1 and interleukin 1 receptor-like 1 [78, 8082]. Moreover, ALK-1 signaling stimulated by TGF-β requires this cytokine to initially activate TβR-II and ALK-5, which then recruit and activate ALK-1 following its association with TβR-II:ALK-5:TGF-β ternary complexes [78]. Thus, activation of ALK-1 and the induction of angiogenesis by TGF-β must first proceed through its assembly of angiostatic TGF-β receptor complexes (i.e., TβR-II:ALK-5). At present, the molecular mechanisms that initially exclude and then recruit ALK-1 to angiostatic TGF-β receptor complexes remain unknown, but may reflect a delicate balance between TGF-β and other angiogenic factors located within tumor microenvironments. Indeed, low TGF-β concentrations enhance the ability of bFGF and VEGF to stimulate EC proliferation and angiogenic sprouting, while high TGF-β concentrations inhibit these angiogenic activities [62, 83]. Along these lines, the pro-angiogenic functions of TGF-β also have been linked to its ability to regulate the expression and/or activities of other angiogenic factors, such as bFGF and VEGF [84]. It is interesting to note that inclusion of TGF-β to Matrigel plugs implanted into mice only promotes angiogenesis and vessel development in the presence of bFGF and its ability to create a pro-angiogenic microenvironment (M. Tian and W.P. Schiemann, unpublished observation). Thus, it is plausible that the recruitment of ALK-1 to angiostatic TGF-β receptor complexes may first require the stimulation of accessory angiogenic signals or proteins within activated EC microenvironments. Along these lines, the coupling of TGF-β to angiogenesis is controlled by the presence of endoglin, whose expression is induced by ALK1 and serves to promote EC proliferation, migration, and tubulogenesis by antagonizing the activities of TβR-I/ALK5 [60, 82].

Collectively, these studies highlight the complexities associated with the ability of TGF-β to regulate EC activities coupled to angiogenesis. Future studies clearly need to (i) better define the precise mechanisms that enable TGF-β and its downstream effectors to govern the induction of angiogenic or angiostatic gene expression profiles; (ii) establish the impact of EC and perivascular cell differentiation states to influence the angiogenic response to TGF-β; and (iii) identify the microenvironmental cues and signals the cooperate with TGF-β in mediating angiogenesis activation and resolution.

2. TGF-β, EMT, and Metastasis

The acquisition of invasive and metastatic phenotypes by carcinomas ushers in their transition from indolent to aggressive disease states, during which time immotile, polarized epithelial cells undergo EMT and transdifferentiate into highly motile, apolar fibroblastoid-like cells [8587]. In doing so, post-EMT carcinoma cells remodel their ECM and microenvironments in a manner that facilitates their intravasation into the vascular or lymphatic systems, as well as their extravasation at distant locales to form micrometastases that ultimately develop into secondary carcinomas [88]. Interestingly, a recent study identified a set of potential metastatic gene signature whose expression is highly associated with the acquisition of pulmonary metastasis by human breast cancers [89]. Included in this metastatic gene signatures are IDs (Inhibitor of Differentiation) 1 and 3, which mediate constitutive proliferative signals in newly established pulmonary micrometastases [89]. In addition, the ability of TGF-β to induce ANGPTL4 (angiopoietin-like 4) expression in breast cancer cells enables their retention, extravasation, and colonization specifically in the lungs, not the bone [90]. Pathological reactivation of EMT programs in differentiated cells and tissues not only promotes their invasion and metastasis, but also underlies the development of several human pathologies, such as chronic inflammation, rheumatoid arthritis, and chronic fibrotic degenerative disorders, all of which are characterized by dysregulated microenvironmental signaling [8588, 91, 92]. In the following sections, we summarize recent developments linking TGF-β to the induction of EMT and metastasis, to the selection and expansion of cancer stem cells, and to the regulation of microRNA expression in developing and progressing neoplasms.

2.1. TGF-β Signaling and EMT

2.1.1. Canonical TGF-β Effectors and EMT

The ability of TGF-β to induce EMT and metastasis transpires through the activation of canonical (i.e., Smad2/3-dependent) and noncanonical (i.e., Smad2/3-independent) TGF-β signaling inputs. For instance, inactivating canonical TGF-β signaling in human MCF10ACA1a breast cancer cells by engineering their expression of a dominant-negative Smad3 construct [93] or a TβR-I mutant incapable of activating Smad2/3 (i.e., L45 mutant) [94] significantly reduced their ability to colonize the lung. Along these lines, Smad4-deficiency not only diminished the expression of PTHrP, IL-11, and CTGF in human MDA-MB-231 breast cancer cells, but also abrogated their metastasis to bone in response to TGF-β [9598]. Interestingly, whereas Smad4-deficiency cooperates with oncogenic K-Ras to induce the initiation and development of pancreatic cancer, the expression and activity of Smad4 is essential for TGF-β stimulation of pancreatic cancer EMT and growth [99]. Similar inactivation of canonical TGF-β signaling by overexpression of Smad7 [100, 101] prevents the invasion of breast [102] and head and neck cancers [103, 104], as well as the pulmonary metastasis of melanomas [105]. Collectively, these findings highlight the importance of Smad2/3/4 signaling in mediating EMT and metastasis stimulated by TGF-β, and suggest the potential benefit of Smad2/3 antagonists to improve the clinical course of patients with metastatic disease.

2.1.2. Noncanonical TGF-β Effectors and EMT

Noncanonical TGF-β signaling also plays an essential role in mediating TGF-β stimulation of EMT, invasion, and metastasis [106]. Included in this growing list of noncanonical effectors targeted by TGF-β are Ras/MAP kinase [107115], PI3K/AKT [116], Rho/ROCK [117], Jagged/Notch [118], mTOR [119], and Wnt/β-catenin [120]. Collaborative signaling events occurring between NF-κB and oncogenic Ras also mediate EMT and pulmonary extravasation of breast cancer cells in response to TGF-β [121]. Similarly, we identified TGF-β stimulation of NF-κB as an essential pathway operant in coupling TGF-β to the expression of Cox-2, whose activity and production of PGE2 are critical for EMT induced by TGF-β in normal and malignant MECs [122]. We [2325] and others [108] also established integrins as key players in mediating EMT, invasion, and p38 MAPK activation by TGF-β, as well as its ability to stimulate the growth and pulmonary metastasis of breast cancers in mice [25]. Essential effectors targeted by the formation of integrin:TGF-β receptor signaling complexes are (i) the protein proto-oncogene Src and its phosphorylation of TβR-II at Y284, which creates a docking site for Grb2 and ShcA [2325]; (ii) the adapter molecule Dab2, which facilitates TGF-β stimulation of Smad2/3 and FAK [123, 124]; and (iii) the protein tyrosine kinase FAK, which coordinates the formation of αvβ3 integrin:TβR-II complexes and, together with its effector p130Cas, is essential for TGF-β stimulation of breast cancer pulmonary metastasis in mice (M.K. Wendt and W.P. Schiemann, unpublished observation). In addition, αvβ3 integrin also mediates TGF-β-dependent metastasis of breast cancer cells to bone [125, 126]. Collectively, these findings implicate TβR-II as an essential mediator of oncogenic signaling by TGF-β, particularly its ability to promote the acquisition of invasive and metastatic phenotypes at the expense of significantly impacting primary tumor growth [127]. Along these lines, a missense mutation in TβR-II identified in human head and neck carcinomas was observed to promote their EMT and invasion in part via (i) hyperactive protein kinase activity in mutant TβR-II proteins, and (ii) inappropriate coupling of TGF-β receptors to Smad1/5 activation, as opposed to Smad2/3 [128]. Interestingly, following its phosphorylation by TβR-II, the tight-junction assembly protein, PAR-6, associates with TβR-I and coordinates the ubiquitination and degradation of RhoA by Smurf1 [129]. The net effect of these TGF-β-dependent events results in the dissolution of epithelial cell tight junctions and the disassembly of their actin cytoskeleton, leading to the induction of EMT.

2.2. TGF-β and Cancer Stem Cells

It is important to note that EMT is a normal and essential physiological process that directs tissue development and morphogenesis in the embryo, as well as promotes the healing, remodeling, and repair of injured tissues in adults [8587]. Thus, tumorigenic EMT in many respects reflects the inappropriate reactivation of embryonic and morphologic gene expression programs, and as such, points towards a potential link between EMT and the maintenance of stem cell properties. Accordingly, aggressive and poorly differentiated breast cancer and glioma cells exhibit gene signatures characteristic of stem cells [130], while human and mouse MECs induced to undergo EMT acquire stem cell-like properties in part via activation of the TGF-β signaling system [131]. Because TGF-β is a master regulator of physiological and pathological EMT [91], these findings suggest that the conversion of TGF-β from a tumor suppressor to a tumor promoter mirrors its ability to induce the selection and expansion of stem cell-like progenitors in post-EMT cells. In fact, TGF-β treatment of malignant, but nonmetastatic human breast cancer cells suppressed their tumorigenicity by diminishing the size of the cancer stem cell pool, and by reducing ID1 expression that results in the differentiation of the progenitor pool [132]. Thus, uncoupling TGF-β from regulation of ID1 expression may dictate whether TGF-β either promotes or suppresses the maintenance and/or expansion of cancer stem cells. Indeed, pharmacological inhibition of TGF-β signaling in cancer stem cells induced a mesenchymal-epithelial transition that resulted in their acquisition of a more epithelial-like morphology [133]. Along these lines, Future studies clearly need to (i) identify the molecular mechanisms that link TGF-β and EMT to the generation of cancer stem cells, and (ii) establish the therapeutic impact of TGF-β in promoting chemoresistance via its stimulation of EMT and the expansion of cancer stem cells.

2.3. TGF-β and microRNAs

Finally, accumulating evidence now positions microRNAs as potentially important regulators of the “TGF-β Paradox.” Indeed, expression of miR-21 in breast cancers predicts for elevated TGF-β1 expression and a poor clinical prognosis [134], while that in gliomas results in the suppression of multiple components of the TGF-β signaling system, including its ligands (e.g., TGF-βs 1 and 3), its receptors (e.g., TβR-II and TβR-III), and its effector molecules (e.g., Smad3, Daxx, and PDCD4) [135, 136]. Recently, TGF-β was shown to promote contractile phenotypes in vascular smooth muscle cells by stimulating the processing of primary miR-21 transcripts into their pre-miR-21 counterparts via the formation of Smad2/3:DROSHA complexes. In doing so, cellular levels of miR-21 accumulate rapidly, resulting in diminished expression of PDCD4 (programmed cell death 4) and its inability to suppress contractile machinery expression in vascular smooth muscle cells [136]. Similar induction of miR-21 expression took place in Smad4-deficient carcinoma cells, suggesting that TGF-β-regulated miR processing also takes place in epithelial cells in a manner independent of Smad4 [136]. Moreover, miR-21 expression also functions to promote EMT stimulated by TGF-β [137], although the molecular mechanisms underlying this event remain to be determined definitively. In contrast to miR-21 and its role in promoting EMT by TGF-β, microRNA-200 family members and miR-205 function in maintaining epithelial cell polarity and, consequently, in suppressing EMT. Importantly, the ability of TGF-β to induce EMT first requires this cytokine to downregulate microRNA-200 family member and miR-205 expression, which promotes ZEB1 and ZEB2 expression and their initiation of EMT [138]. Thus, aberrant microRNA expression may play a significant role in determining whether epithelial cells sense and respond to the tumor suppressing functions of TGF-β, or rather to its oncogenic activities.

Conclusions and Future Perspectives

Despite considerable progress over the last decade in defining the molecular mechanisms that underlie the initiation and maintenance of the “TGF-β Paradox,” science and medicine still lack the necessary knowledge and wherewithal to explain and, more importantly, to manipulate the physiopathological actions of TGF-β to improve the clinical course of human malignancies. While it is abundantly clear that TGF-β plays a major role, both directly and indirectly, in regulating the ability of cancer cells to acquire each of the 6 hallmarks necessary for their malignant progression [139], it remains unclear as to how these events conspire in regulating the response of developing and progressing neoplasms to TGF-β. For instance, defects in TGF-β function rarely effect primary tumor growth, but more commonly play a significant role in enabling cancer cells to acquire EMT and invasive/metastatic phenotypes. Thus, while it is easy to rationalize why tumors require TGF-β to provide them with a selective EMT and metastatic advantage, teleologically it remains troublesome to assume that these phenotypic changes induced by TGF-β are permanently ingrained in aggressive carcinoma cells. Indeed, cancer cells perpetually locked into a “vagabond” mentally is counterintuitive to the processes underlying metastasis development and the formation of secondary carcinomas at distant locales. Instead, it appears that the exquisite balance between the functions and behaviors of TGF-β in distinct tissue types become unbalanced and incapable of suppressing disease development, particularly that of neoplastic transformation. Along these lines, the processes underlying the maintenance of normal tissue and cellular homeostasis have been liken to those necessary in facilitating the existence of a well-balanced and harmonious society [140]. The studies highlighted herein are consistent with a role for TGF-β in serving either as a benevolent or corrupt village manager, one whose ultimate agenda is dictated by the prevailing mood of the village’s stromal and microenvironmental constituents. Thus, these findings also underscore and reinforce the need to develop novel pharmacological agents designed to antagonize the oncogenic activities of TGF-β in cancer cells, as well as in their supporting stromal compartments.

Executive Summary

TGF-β and the Tumor Microenvironment Fibroblasts

TGF-β and Immunosurveillance

TGF-β and Endothelial Cells

TGF-β, EMT, and Metastasis

Acknowledgements

Members of the Schiemann Laboratory are thanked for critical comments and reading of the manuscript. W.P.S. was supported by grants from the National Institutes of Health (CA114039 and CA129359), the Komen Foundation (BCTR0706967), and the Department of Defense (BC084651).

References

1. Bierie B, Moses HL. Tumour microenvironment: TGF-β: the molecular Jekyll and Hyde of cancer. Nat Rev Cancer. 2006;6:506–520. [PubMed] [Google Scholar]

2. Mueller MM, Fusenig NE. Friends or foes - bipolar effects of the tumour stroma in cancer. Nat. Rev. Cancer. 2004;4:839–849. [PubMed] [Google Scholar]

3. Bhowmick NA, Chytil A, Plieth D, et al. TGF-β signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science. 2004;303:848–851. [PubMed] [Google Scholar]

5. Bhowmick NA, Neilson EG, Moses HL. Stromal fibroblasts in cancer initiation and progression. Nature. 2004;432:332–337. [PMC free article] [PubMed] [Google Scholar]

6. Fidler IJ. Critical determinants of metastasis. Semin. Cancer Biol. 2002;12:89–96. [PubMed] [Google Scholar]

7. Fidler IJ. The organ microenvironment and cancer metastasis. Differentiation. 2002;70:498–505. [PubMed] [Google Scholar]

8. Cheng N, Bhowmick NA, Chytil A, et al. Loss of TGF-β type II receptor in fibroblasts promotes mammary carcinoma growth and invasion through upregulation of TGF-α-, MSP-and HGF-mediated signaling networks. Oncogene. 2005;24:5053–5068. [PMC free article] [PubMed] [Google Scholar]

9. Yang L, Huang J, Ren X, et al. Abrogation of TGF-β signaling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell. 2008;13:23–35. [PMC free article] [PubMed] [Google Scholar]

10. Bruna A, Darken RS, Rojo F, et al. High TGF-β-Smad activity confers poor prognosis in glioma patients and promotes cell proliferation depending on the methylation of the PDGF-B gene. Cancer Cell. 2007;11:147–160. [PubMed] [Google Scholar]

11. Paszek MJ, Weaver VM. The tension mounts: mechanics meets morphogenesis and malignancy. J. Mammary Gland Biol. Neoplasia. 2004;9:325–342. [PubMed] [Google Scholar]

12. Anderson AR, Weaver AM, Cummings PT, Quaranta V. Tumor morphology and phenotypic evolution driven by selective pressure from the microenvironment. Cell. 2006;127:905–915. [PubMed] [Google Scholar]

13. Lucero HA, Kagan HM. Lysyl oxidase: an oxidative enzyme and effector of cell function. Cell. Mol. Life Sci. 2006;63:2304–2316. [PMC free article] [PubMed] [Google Scholar]

14. Payne SL, Hendrix MJ, Kirschmann DA. Paradoxical roles for lysyl oxidases in cancer--a prospect. J. Cell. Biochem. 2007;101:1338–1354. [PubMed] [Google Scholar]

15. Erler JT, Giaccia AJ. Lysyl oxidase mediates hypoxic control of metastasis. Cancer Res. 2006;66:10238–10241. [PubMed] [Google Scholar]

16. Kagan HM, Li W. Lysyl oxidase: properties, specificity, and biological roles inside and outside of the cell. J. Cell. Biochem. 2003;88:660–672. [PubMed] [Google Scholar]

17. Atsawasuwan P, Mochida Y, Katafuchi M, et al. Lysyl oxidase binds TGF-β and regulates its signaling via amine oxidase activity. J. Biol. Chem. 2008;283:34229–34240. [PMC free article] [PubMed] [Google Scholar]

18. Kim DJ, Lee DC, Yang SJ, et al. Lysyl oxidase like 4, a novel target gene of TGF-β1 signaling, can negatively regulate TGF-beta1-induced cell motility in PLC/PRF/5 hepatoma cells. Biochem. Biophys. Res. Commun. 2008;373:521–527. [PubMed] [Google Scholar]

19. Erler JT, Bennewith KL, Nicolau M, et al. Lysyl oxidase is essential for hypoxia-induced metastasis. Nature. 2006;440:1222–1226. [PubMed] [Google Scholar]

20. Kirschmann DA, Seftor EA, Fong SF, et al. A molecular role for lysyl oxidase in breast cancer invasion. Cancer Res. 2002;62:4478–4483. [PubMed] [Google Scholar]

21. Payne SL, Fogelgren B, Hess AR, et al. Lysyl oxidase regulates breast cancer cell migration and adhesion through a hydrogen peroxide-mediated mechanism. Cancer Res. 2005;65:11429–11436. [PubMed] [Google Scholar]

22. Payne SL, Hendrix MJ, Kirschmann DA. Lysyl oxidase regulates actin filament formation through the p130(Cas)/Crk/DOCK180 signaling complex. J. Cell. Biochem. 2006;98:827–837. [PubMed] [Google Scholar]

23. Galliher AJ, Schiemann WP. β3 integrin and Src facilitate TGF-β mediated induction of epithelial-mesenchymal transition in mammary epithelial cells. Breast Cancer Res. 2006;8:R42. [PMC free article] [PubMed] [Google Scholar]

24. Galliher AJ, Schiemann WP. Src phosphorylates Tyr284 in TGF-β type II receptor and regulates TGF-β stimulation of p38 MAPK during breast cancer cell proliferation and invasion. Cancer Res. 2007;67:3752–3758. [PubMed] [Google Scholar]

25. Galliher-Beckley AJ, Schiemann WP. Grb2 Binding to Tyr284 in TβR-II is essential for mammary tumor growth and metastasis stimulated by TGF-β Carcinogenesis. 2008;29:244–251. [PMC free article] [PubMed] [Google Scholar]

26. Andarawewa KL, Paupert J, Pal A, Barcellos-Hoff MH. New rationales for using TGF-β inhibitors in radiotherapy. Int. J. Radiat. Biol. 2007;83:803–811. [PubMed] [Google Scholar]

27. Barcellos-Hoff MH, Medina D. New highlights on stroma-epithelial interactions in breast cancer. Breast Cancer Res. 2005;7:33–36. [PMC free article] [PubMed] [Google Scholar]

28. Biswas S, Guix M, Rinehart C, et al. Inhibition of TGF-β with neutralizing antibodies prevents radiation-induced acceleration of metastatic cancer progression. J. Clin. Invest. 2007;117:1305–1313. [PMC free article] [PubMed] [Google Scholar]

29. Lin WW, Karin M. A cytokine-mediated link between innate immunity, inflammation, and cancer. J. Clin. Invest. 2007;117:1175–1183. [PMC free article] [PubMed] [Google Scholar]

30. Li MO, Wan YY, Sanjabi S, Robertson AK, Flavell RA. TGF-β regulation of immune responses. Annu. Rev. Immunol. 2006;24:99–146. [PubMed] [Google Scholar]

31. Gorelik L, Flavell RA. Abrogation of TGF-β signaling in T cells leads to spontaneous T cell differentiation and autoimmune disease. Immunity. 2000;12:171–181. [PubMed] [Google Scholar]

32. Kulkarni AB, Huh CG, Becker D, et al. TGF-β1 null mutation in mice causes excessive inflammatory response and early death. Proc. Natl. Acad. Sci. U.S.A. 1993;90:770–774. [PMC free article] [PubMed] [Google Scholar]

33. Yang X, Letterio JJ, Lechleider RJ, et al. Targeted disruption of SMAD3 results in impaired mucosal immunity and diminished T cell responsiveness to TGF-β EMBO J. 1999;18:1280–1291. [PMC free article] [PubMed] [Google Scholar]

34. Kim BG, Li C, Qiao W, et al. Smad4 signalling in T cells is required for suppression of gastrointestinal cancer. Nature. 2006;441:1015–1019. [PubMed] [Google Scholar]

35. Dalal BI, Keown PA, Greenberg AH. Immunohistochemical localization of secreted TGF-β1 to the advancing edges of primary tumors and to lymph node metastases of human mammary carcinoma. Am. J. Pathol. 1993;143:381–389. [PMC free article] [PubMed] [Google Scholar]

36. Gorsch SM, Memoli VA, Stukel TA, Gold LI, Arrick BA. Immunohistochemical staining for TGF-β1 associates with disease progression in human breast cancer. Cancer Res. 1992;52:6949–6952. [PubMed] [Google Scholar]

37. Ivanovic V, Todorovic-Rakovic N, Demajo M, et al. Elevated plasma levels of TGF-β1 in patients with advanced breast cancer: Association with disease progression. Eur. J. Cancer. 2003;39:454–461. [PubMed] [Google Scholar]

38. Lin SJ, Chang C, Ng AK, Wang SH, Li JJ, Hu CP. Prevention of TGF-β-induced apoptosis by interlukin-4 through Akt activation and p70S6K survival signaling pathways. Apoptosis. 2007;12:1659–1670. [PubMed] [Google Scholar]

39. Teicher BA. TGF-β and the immune response to malignant disease. Clin. Cancer Res. 2007;13:6247–6251. [PubMed] [Google Scholar]

40. Wrzesinski SH, Wan YY, Flavell RA. TGF-β and the immune response: implications for anticancer therapy. Clin. Cancer Res. 2007;13:5262–5270. [PubMed] [Google Scholar]

41. Chen Ml, Pittet MJ, Gorelik L, et al. Regulatory T cells suppress tumor-specific CD8 T cell cytotoxicity through TGF-β signals in vivo. Cancer Res. 2006;66:8617–8624. [PMC free article] [PubMed] [Google Scholar]

42. Gorelik L, Flavell RA. TGF-β in T-cell biology. Nat. Rev. Immunol. 2002;2:46–53. [PubMed] [Google Scholar]

43. Gorelik L, Flavell RA. Immune-mediated eradication of tumors through the blockade of TGF-β signaling in T cells. Nat. Med. 2001;7:1118–1122. [PubMed] [Google Scholar]

44. Zhang Q, Yang X, Pins M, et al. Adoptive transfer of tumor-reactive TGF-β-insensitive CD8+ T cells: Eradication of autologous mouse prostate cancer. Cancer Res. 2005;65:1761–1769. [PubMed] [Google Scholar]

45. Nam JS, Terabe M, Kang MJ, et al. TGF-β subverts the immune system into directly promoting tumor growth through interleukin-17. Cancer Res. 2008;68:3915–3923. [PMC free article] [PubMed] [Google Scholar]

46. Neil JR, Schiemann WP. Altered TAB1:IκB kinase interaction promotes TGF-β-mediated NF-κB activation during breast cancer progression. Cancer Res. 2008;68:1462–1470. [PMC free article] [PubMed] [Google Scholar]

47. Lu T, Tian L, Han Y, Vogelbaum M, Stark GR. Dose-dependent cross-talk between the TGF-β and interleukin-1 signaling pathways. Proc. Natl. Acad. Sci. U.S.A. 2007;104:4365–4370. [PMC free article] [PubMed] [Google Scholar]

48. Fries G, Perneczky A, Kempski O. Enhanced interleukin-1β release and longevity of glioma-associated peripheral blood monocytes in vitro. Neurosurgery. 1994;35:264–270. [PubMed] [Google Scholar]

49. Griffin BD, Moynagh PN. Persistent interleukin-1β signaling causes long term activation of NF-κB in a promoter-specific manner in human glial cells. J. Biol. Chem. 2006;281:10316–10326. [PubMed] [Google Scholar]

50. Tu S, Bhagat G, Cui G, et al. Overexpression of interleukin-1β induces gastric inflammation and cancer and mobilizes myeloid-derived suppressor cells in mice. Cancer Cell. 2008;14:408–419. [PMC free article] [PubMed] [Google Scholar]

51. Larmonier N, Cathelin D, Larmonier C, et al. The inhibition of TNF-α anti-tumoral properties by blocking antibodies promotes tumor growth in a rat model. Exp. Cell Res. 2007;313:2345–2355. [PubMed] [Google Scholar]

52. Conti P, Castellani MI, Kempuraj D, et al. Role of mast cells in tumor growth. Ann. Clin. Lab. Sci. 2007;37:315–321. [PubMed] [Google Scholar]

53. Lin EY, Li JF, Gnatovskiy L, et al. Macrophages regulate the angiogenic switch in a mouse model of breast cancer. Cancer Res. 2006;66:11238–11246. [PubMed] [Google Scholar]

54. Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat. Rev. Cancer. 2004;4:71–78. [PubMed] [Google Scholar]

55. Carmeliet P. Mechanisms of angiogenesis and arteriogenesis. Nat. Med. 2000;6:389–395. [PubMed] [Google Scholar]

56. Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000;407:249–257. [PubMed] [Google Scholar]

57. Carmeliet P. Angiogenesis in health and disease. Nat. Med. 2003;9:653–660. [PubMed] [Google Scholar]

58. Erler JT, Weaver VM. Three-dimensional context regulation of metastasis. Clin. Exp. Metastasis. 2008 PMID: 18814043. [PMC free article] [PubMed] [Google Scholar]

59. Bertolino P, Deckers M, Lebrin F, ten Dijke P. TGF-β signal transduction in angiogenesis and vascular disorders. Chest. 2005;128:585S–590S. [PubMed] [Google Scholar]

60. Lebrin F, Deckers M, Bertolino P, Ten Dijke P. TGF-β receptor function in the endothelium. Cardiovasc. Res. 2005;65:599–608. [PubMed] [Google Scholar]

61. Pepper MS. TGF-β: vasculogenesis, angiogenesis, and vessel wall integrity. Cytokine Growth Factor Rev. 1997;8:21–43. [PubMed] [Google Scholar]

62. Pepper MS, Vassalli JD, Orci L, Montesano R. Biphasic effect of TGF-β1 on in vitro angiogenesis. Exp. Cell Res. 1993;204:356–363. [PubMed] [Google Scholar]

63. Dickson MC, Martin JS, Cousins FM, Kulkarni AB, Karlsson S, Akhurst RJ. Defective haematopoiesis and vasculogenesis in TGF-β1 knock out mice. Development. 1995;121:1845–1854. [PubMed] [Google Scholar]

64. Larsson J, Goumans MJ, Sjostrand LJ, et al. Abnormal angiogenesis but intact hematopoietic potential in TGF-β type I receptor-deficient mice. EMBO J. 2001;20:1663–1673. [PMC free article] [PubMed] [Google Scholar]

65. Goumans MJ, Zwijsen A, van Rooijen MA, Huylebroeck D, Roelen BA, Mummery CL. TGF-β signalling in extraembryonic mesoderm is required for yolk sac vasculogenesis in mice. Development. 1999;126:3473–3483. [PubMed] [Google Scholar]

66. Oshima M, Oshima H, Taketo MM. TGF-β receptor type II deficiency results in defects of yolk sac hematopoiesis and vasculogenesis. Dev. Biol. 1996;179:297–302. [PubMed] [Google Scholar]

67. Compton LA, Potash DA, Brown CB, Barnett JV. Coronary vessel development is dependent on the type III TGF-β receptor. Circ. Res. 2007;101:784–791. [PubMed] [Google Scholar]

68. Brown CB, Boyer AS, Runyan RB, Barnett JV. Requirement of type III TGF-β receptor for endocardial cell transformation in the heart. Science. 1999;283:2080–2082. [PubMed] [Google Scholar]

69. Lechleider RJ, Ryan JL, Garrett L, et al. Targeted mutagenesis of Smad1 reveals an essential role in chorioallantoic fusion. Dev. Biol. 2001;240:157–167. [PubMed] [Google Scholar]

70. Chang H, Huylebroeck D, Verschueren K, Guo Q, Matzuk MM, Zwijsen A. Smad5 knockout mice die at mid-gestation due to multiple embryonic and extraembryonic defects. Development. 1999;126:1631–1642. [PubMed] [Google Scholar]

71. McAllister KA, Grogg KM, Johnson DW, et al. Endoglin, a TGF-β binding protein of endothelial cells, is the gene for hereditary haemorrhagic telangiectasia type 1. Nat. Genet. 1994;8:345–351. [PubMed] [Google Scholar]

72. Shovlin CL, Hughes JM, Scott J, Seidman CE, Seidman JG. Characterization of endoglin and identification of novel mutations in hereditary hemorrhagic telangiectasia. Am. J. Hum. Genet. 1997;61:68–79. [PMC free article] [PubMed] [Google Scholar]

73. Berg JN, Gallione CJ, Stenzel TT, et al. The activin receptor-like kinase 1 gene: Genomic structure and mutations in hereditary hemorrhagic telangiectasia type 2. Am. J. Hum. Genet. 1997;61:60–67. [PMC free article] [PubMed] [Google Scholar]

74. Johnson DW, Berg JN, Baldwin MA, et al. Mutations in the activin receptor-like kinase 1 gene in hereditary haemorrhagic telangiectasia type 2. Nat. Genet. 1996;13:189–195. [PubMed] [Google Scholar]

75. Arthur HM, Ure J, Smith AJ, et al. Endoglin, an ancillary TGF-β receptor, is required for extraembryonic angiogenesis and plays a key role in heart development. Dev. Biol. 2000;217:42–53. [PubMed] [Google Scholar]

76. Bourdeau A, Dumont DJ, Letarte M. A murine model of hereditary hemorrhagic telangiectasia. J. Clin. Invest. 1999;104:1343–1351. [PMC free article] [PubMed] [Google Scholar]

77. Srinivasan S, Hanes MA, Dickens T, et al. A mouse model for hereditary hemorrhagic telangiectasia (HHT) type 2. Hum. Mol. Genet. 2003;12:473–482. [PubMed] [Google Scholar]

78. Goumans MJ, Valdimarsdottir G, Itoh S, et al. Activin receptor-like kinase (ALK)1 is an antagonistic mediator of lateral TGF-β/ALK5 signaling. Mol. Cell. 2003;12:817–828. [PubMed] [Google Scholar]

79. Oh SP, Seki T, Goss KA, et al. Activin receptor-like kinase 1 modulates TGF-β1 signaling in the regulation of angiogenesis. Proc. Natl. Acad. Sci. U.S.A. 2000;97:2626–2631. [PMC free article] [PubMed] [Google Scholar]

80. Goumans MJ, Valdimarsdottir G, Itoh S, Rosendahl A, Sideras P, ten Dijke P. Balancing the activation state of the endothelium via two distinct TGF-β type I receptors. EMBO J. 2002;21:1743–1753. [PMC free article] [PubMed] [Google Scholar]

81. Wu X, Ma J, Han JD, Wang N, Chen YG. Distinct regulation of gene expression in human endothelial cells by TGF-β and its receptors. Microvasc. Res. 2006;71:12–19. [PubMed] [Google Scholar]

82. Ota T, Fujii M, Sugizaki T, et al. Targets of transcriptional regulation by two distinct type I receptors for TGF-β in human umbilical vein endothelial cells. J. Cell Physiol. 2002;193:299–318. [PubMed] [Google Scholar]

83. Fajardo LF, Prionas SD, Kwan HH, Kowalski J, Allison AC. TGF-β1 induces angiogenesis in vivo with a threshold pattern. Lab. Invest. 1996;74:600–608. [PubMed] [Google Scholar]

84. Goumans MJ, Lebrin F, Valdimarsdottir G. Controlling the angiogenic switch: a balance between two distinct TGF-β receptor signaling pathways. Trends Cardiovasc. Med. 2003;13:301–307. [PubMed] [Google Scholar]

85. Savagner P. Leaving the neighborhood: Molecular mechanisms involved during epithelial-mesenchymal transition. Bioessays. 2001;23:912–923. [PubMed] [Google Scholar]

86. Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat. Rev. Cancer. 2002;2:442–454. [PubMed] [Google Scholar]

87. Thiery JP. Epithelial-mesenchymal transitions in development and pathologies. Curr. Opin. Cell Biol. 2003;15:740–746. [PubMed] [Google Scholar]

88. Zavadil J, Bottinger EP. TGF-β and epithelial-to-mesenchymal transitions. Oncogene. 2005;24:5764–5774. [PubMed] [Google Scholar]

89. Gupta GP, Perk J, Acharyya S, et al. ID genes mediate tumor reinitiation during breast cancer lung metastasis. Proc. Natl. Acad. Sci. U.S.A. 2007;104:19506–19511. [PMC free article] [PubMed] [Google Scholar]

90. Padua D, Zhang XH, Wang Q, et al. TGF-β primes breast tumors for lung metastasis seeding through angiopoietin-like 4. Cell. 2008;133:66–77. [PMC free article] [PubMed] [Google Scholar]

91. Moustakas A, Heldin CH. Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression. Cancer Sci. 2007;98:1512–1520. [PMC free article] [PubMed] [Google Scholar]

92. Willis BC, Borok Z. TGF-β-induced EMT: Mechanisms and implications for fibrotic lung disease. Am. J. Physiol. Lung Cell. Mol. Physiol. 2007;293:L525–L534. [PubMed] [Google Scholar]

93. Tian F, DaCosta Byfield S, Parks WT, et al. Reduction in Smad2/3 signaling enhances tumorigenesis but suppresses metastasis of breast cancer cell lines. Cancer Res. 2003;63:8284–8292. [PubMed] [Google Scholar]

94. Tian F, Byfield SD, Parks WT, et al. Smad-binding defective mutant of TGF-β type I receptor enhances tumorigenesis but suppresses metastasis of breast cancer cell lines. Cancer Res. 2004;64:4523–4530. [PubMed] [Google Scholar]

95. Kang Y, He W, Tulley S, et al. Breast cancer bone metastasis mediated by the Smad tumor suppressor pathway. Proc. Natl. Acad. Sci. U.S.A. 2005;102:13909–13914. [PMC free article] [PubMed] [Google Scholar]

96. Kang Y, Siegel PM, Shu W, et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell. 2003;3:537–549. [PubMed] [Google Scholar]

97. Yin JJ, Selander K, Chirgwin JM, et al. TGF-β signaling blockade inhibits PTHrP secretion by breast cancer cells and bone metastases development. J. Clin. Invest. 1999;103:197–206. [PMC free article] [PubMed] [Google Scholar]

98. Deckers M, van Dinther M, Buijs J, et al. The tumor suppressor Smad4 is required for TGF-β-induced epithelial to mesenchymal transition and bone metastasis of breast cancer cells. Cancer Res. 2006;66:2202–2209. [PubMed] [Google Scholar]

99. Bardeesy N, Cheng KH, Berger JH, et al. Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes Dev. 2006;20:3130–3146. [PMC free article] [PubMed] [Google Scholar]

100. Hayashi H, Abdollah S, Qiu Y, et al. The MAD-related protein Smad7 associates with the TGF-β receptor and functions as an antagonist of TGF-β signaling. Cell. 1997;89:1165–1173. [PubMed] [Google Scholar]

101. Souchelnytskyi S, Nakayama T, Nakao A, et al. Physical and functional interaction of murine and Xenopus Smad7 with bone morphogenetic protein receptors and TGF-β receptors. J. Biol. Chem. 1998;273:25364–25370. [PubMed] [Google Scholar]

102. Azuma H, Ehata S, Miyazaki H, et al. Effect of Smad7 expression on metastasis of mouse mammary carcinoma JygMC(A) cells. J. Natl. Cancer Inst. 2005;97:1734–1746. [PubMed] [Google Scholar]

103. Leivonen SK, Ala-Aho R, Koli K, Grenman R, Peltonen J, Kahari VM. Activation of Smad signaling enhances collagenase-3 (MMP-13) expression and invasion of head and neck squamous carcinoma cells. Oncogene. 2006;25:2588–2600. [PubMed] [Google Scholar]

104. Leivonen SK, Kahari VM. TGF-β signaling in cancer invasion and metastasis. Int. J. Cancer. 2007;121:2119–2124. [PubMed] [Google Scholar]

105. Javelaud D, Mohammad KS, McKenna CR, et al. Stable overexpression of Smad7 in human melanoma cells impairs bone metastasis. Cancer Res. 2007;67:2317–2324. [PubMed] [Google Scholar]

106. Moustakas A, Heldin CH. Non-Smad TGF-β signals. J. Cell Sci. 2005;118:3573–3584. [PubMed] [Google Scholar]

107. Bakin AV, Rinehart C, Tomlinson AK, Arteaga CL. p38 mitogen-activated protein kinase is required for TGF-β-mediated fibroblastic transdifferentiation and cell migration. J. Cell Sci. 2002;115:3193–3206. [PubMed] [Google Scholar]

108. Bhowmick NA, Zent R, Ghiassi M, McDonnell M, Moses HL. Integrin β1 signaling is necessary for TGF-β activation of p38MAPK and epithelial plasticity. J. Biol. Chem. 2001;276:46707–46713. [PubMed] [Google Scholar]

109. Cui W, Fowlis DJ, Bryson S, et al. TGF-β1 inhibits the formation of benign skin tumors, but enhances progression to invasive spindle carcinomas in transgenic mice. Cell. 1996;86:531–542. [PubMed] [Google Scholar]

110. Davies M, Robinson M, Smith E, Huntley S, Prime S, Paterson I. Induction of an epithelial to mesenchymal transition in human immortal and malignant keratinocytes by TGF-β1 involves MAPK, Smad and AP-1 signalling pathways. J. Cell. Biochem. 2005;95:918–931. [PubMed] [Google Scholar]

111. Ellenrieder V, Hendler SF, Boeck W, et al. TGF-β1 treatment leads to an epithelial-mesenchymal transdifferentiation of pancreatic cancer cells requiring extracellular signal-regulated kinase 2 activation. Cancer Res. 2001;61:4222–4228. [PubMed] [Google Scholar]

112. Fowlis DJ, Cui W, Johnson SA, Balmain A, Akhurst RJ. Altered epidermal cell growth control in vivo by inducible expression of TGF-β1 in the skin of transgenic mice. Cell Growth Differ. 1996;7:679–687. [PubMed] [Google Scholar]

113. Janda E, Lehmann K, Killisch I, et al. Ras and TGF-β cooperatively regulate epithelial cell plasticity and metastasis: Dissection of Ras signaling pathways. J. Cell Biol. 2002;156:299–313. [PMC free article] [PubMed] [Google Scholar]

114. Lehmann K, Janda E, Pierreux CE, et al. Raf induces TGF-β production while blocking its apoptotic but not invasive responses: A mechanism leading to increased malignancy in epithelial cells. Genes Dev. 2000;14:2610–2622. [PMC free article] [PubMed] [Google Scholar]

115. Oft M, Akhurst RJ, Balmain A. Metastasis is driven by sequential elevation of H-ras and Smad2 levels. Nat. Cell Biol. 2002;4:487–494. [PubMed] [Google Scholar]

116. Bakin AV, Tomlinson AK, Bhowmick NA, Moses HL, Arteaga CL. Phosphatidylinositol 3-kinase function is required for TGF-β-mediated epithelial to mesenchymal transition and cell migration. J. Biol. Chem. 2000;275:36803–36810. [PubMed] [Google Scholar]

117. Bhowmick NA, Ghiassi M, Bakin A, et al. TGF-β1 mediates epithelial to mesenchymal transdifferentiation through a RhoA-dependent mechanism. Mol. Biol. Cell. 2001;12:27–36. [PMC free article] [PubMed] [Google Scholar]

118. Zavadil J, Cermak L, Soto-Nieves N, Bottinger EP. Integration of TGF-β/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition. EMBO J. 2004;23:1155–1165. [PMC free article] [PubMed] [Google Scholar]

119. Lamouille S, Derynck R. Cell size and invasion in TGF-β-induced epithelial to mesenchymal transition is regulated by activation of the mTOR pathway. J. Cell Biol. 2007;178:437–451. [PMC free article] [PubMed] [Google Scholar]

120. Kim K, Lu Z, Hay ED. Direct evidence for a role of β-catenin/LEF-1 signaling pathway in induction of EMT. Cell Biol. Int. 2002;26:463–476. [PubMed] [Google Scholar]

121. Huber MA, Azoitei N, Baumann B, et al. NF-κB is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression. J. Clin. Invest. 2004;114:569–581. [PMC free article] [PubMed] [Google Scholar]

122. Neil JR, Johnson KM, Nemenoff RA, Schiemann WP. Cox-2 inactivates Smad signaling and enhances EMT stimulated by TGF-β through a PGE2-dependent mechanism. Carcinogenesis. 2008;29:2227–2235. [PMC free article] [PubMed] [Google Scholar]

123. Hocevar BA, Smine A, Xu XX, Howe PH. The adaptor molecule Disabled-2 links the TGF-β receptors to the Smad pathway. EMBO J. 2001;20:2789–2801. [PMC free article] [PubMed] [Google Scholar]

124. Prunier C, Howe PH. Disabled-2 (Dab2) is required for TGF-β-induced epithelial to mesenchymal transition (EMT) J. Biol. Chem. 2005;280:17540–17548. [PubMed] [Google Scholar]

125. Bandyopadhyay A, Agyin JK, Wang L, et al. Inhibition of pulmonary and skeletal metastasis by a TGF-β type I receptor kinase inhibitor. Cancer Res. 2006;66:6714–6721. [PubMed] [Google Scholar]

126. Sloan EK, Pouliot N, Stanley KL, et al. Tumor-specific expression of αvβ3 integrin promotes spontaneous metastasis of breast cancer to bone. Breast Cancer Res. 2006;8:R20. [PMC free article] [PubMed] [Google Scholar]

127. Muraoka-Cook RS, Kurokawa H, Koh Y, et al. Conditional overexpression of active TGF-β1 in vivo accelerates metastases of transgenic mammary tumors. Cancer Res. 2004;64:9002–9011. [PubMed] [Google Scholar]

128. Bharathy S, Xie W, Yingling JM, Reiss M. Cancer-associated TGF-β type II receptor gene mutant causes activation of bone morphogenic protein-Smads and invasive phenotype. Cancer Res. 2008;68:1656–1666. [PMC free article] [PubMed] [Google Scholar]

129. Ozdamar B, Bose R, Barrios-Rodiles M, Wang HR, Zhang Y, Wrana JL. Regulation of the polarity protein Par6 by TGF-β receptors controls epithelial cell plasticity. Science. 2005;307:1603–1609. [PubMed] [Google Scholar]

130. Ben-Porath I, Thomson MW, Carey VJ, et al. An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat. Genet. 2008;40:499–507. [PMC free article] [PubMed] [Google Scholar]

131. Mani SA, Guo W, Liao MJ, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133:704–715. [PMC free article] [PubMed] [Google Scholar]

132. Tang B, Yoo N, Vu M, et al. TGF-β can suppress tumorigenesis through effects on the putative cancer stem or early progenitor cell and committed progeny in a breast cancer xenograft model. Cancer Res. 2007;67:8643–8652. [PMC free article] [PubMed] [Google Scholar]

133. Shipitsin M, Campbell LL, Argani P, et al. Molecular definition of breast tumor heterogeneity. Cancer Cell. 2007;11:259–273. [PubMed] [Google Scholar]

134. Qian B, Katsaros D, Lu L, et al. High miR-21 expression in breast cancer associated with poor disease-free survival in early stage disease and high TGF-β1. Breast Cancer Res. Treat. 2008 PMID: 18932017. [PubMed] [Google Scholar]

135. Papagiannakopoulos T, Shapiro AKosik KS. MicroRNA-21 targets a network of key tumor-suppressive pathways in glioblastoma cells. Cancer Res. 2008;68:8164–8172. [PubMed] [Google Scholar]

136. Davis BN, Hilyard AC, Lagna GHata A. SMAD proteins control DROSHA-mediated microRNA maturation. Nature. 2008;454:56–61. [PMC free article] [PubMed] [Google Scholar]

137. Zavadil J, Narasimhan M, Blumenberg M, Schneider RJ. TGF-β and microRNA:mRNA regulatory networks in epithelial plasticity. Cells Tissues Organs. 2007;185:157–161. [PubMed] [Google Scholar]

138. Gregory PA, Bert AG, Paterson EL, et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat. Cell Biol. 2008;10:593–601. [PubMed] [Google Scholar]

139. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100(1):57–70. [PubMed] [Google Scholar]

140. Rizki A, Bissell MJ. Homeostasis in the breast: It takes a village. Cancer Cell. 2004;6:1–2. [PubMed] [Google Scholar]