首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
TGFβ1 is a multifunctional factor, controlling cellular growth and extracellular matrix production. Deletion of the TGFβ1 gene in mice results in multiple inflammatory reactions. Targeted overexpression of TGFβ1 in pancreatic islet cells leads to fibrosis of the exocrine pancreas in transgenic mice. In pancreatic fibrosis interstitial fibroblasts are primary candidates for production and deposition of extracellular matrix. Still, little is known about regulation of these cells during development of pancreatic disease. We established primary cell lines of pancreatic fibroblastoid/stellate cells (PFC) from rat pancreas. Investigation of rPFCs in vitro shows TGFβ1 expression by RT-PCR analysis. Mature TGFβ1 was detected in culture supernatants by immunoassay. Rat PFCs in culture possess both receptors TGFβ receptor type I, and type II, necessary for TGFβ1 signal transduction. Inhibition of TGFβ1 activity by means of neutralizing antibodies interferes with an autocrine loop and results in a 2-fold stimulation of cell growth. So far, pancreatic fibroblastoid/stellate cells in vitro were known as a target of TGFβ1 action, but not as a source of TGFβ1. Our data indicate TGFβ1 activity in rat pancreas extends beyond regulation of matrix production, but appears to be important in growth control of pancreatic fibroblastoid cells.  相似文献   

2.
Transforming growth factor-β (TGFβ) superfamily members exert their diverse biological effects through their interaction with heteromeric receptor complexes of transmembrane serine/threonine kinases. Both components of the receptor complex, known as receptor I and receptor II are essential for signal transduction. The composition of these complexes can vary significantly due to the promiscuous nature of the ligands and the receptors, and this diversity of interactions can yield a variety of biological responses. Several receptor interacting proteins and potential mediators of signal transduction have now been identified. Recent advances, particularly in our understanding of the function of Mothers against dpp-related (MADR) proteins, are providing new insights into how the TGFβ superfamily signals its diverse biological activities.  相似文献   

3.
4.
5.
6.
Transforming growth factor β1 (TGFβ) inhibits cellular proliferation, promotes differentiation, and stimulates the expression and secretion of the extracellular matrix adhesion molecules fibronectin and laminin and the colon-associated intercellular adhesion molecule carcinoembryonic antigen. This is collectively called the TGFβ-mediated adhesion response and occurs in the human colon cancer cell line Moser while the cell line KM12SM is relatively unresponsive to TGFβ. We have previously shown that TGFβ rapidly stimulates protein kinase C (PKC) phosphotransferase activity in the Moser cells and that the induction of the adhesion response (but not antiproliferation) by TGFβ is dependent on PKC. Because resistance to growth factors may be due to translational suppression and the translation initiation factor eIF-4E may alleviate translational suppression, we determined the effect of eIF-4E expression on the responses of Moser and KM12SM cells to TGFβ. Ectopic expression of eIF-4E in the TGFβ-responsive Moser cells enhanced the activation of PKC by TGFβ and the induction of the adhesion response, especially the secretion of adhesion molecules, but not the antiproliferative response. Ectopic expression of eIF-4E in the TGFβ-resistant KM12SM cells increased TGFβ stimulation of PKC and the TGFβ-mediated adhesion response (but not antiproliferation). The secretion of adhesion molecules was significantly increased by TGFβ. These results showed in these cells that eIF-4E promotes TGFβ-regulated adhesion but not antiproliferation in a PKC-dependent manner.  相似文献   

7.
TGFβ can promote and/or suppress prostate tumor growth through multiple and opposing actions. Alterations of its expression, secretion, regulation or of the sensitivity of target cells can lead to a favorable environment for tumor development. To gain a better insight in TGFβ function during cancer progression, we have used different cultured human prostate cells: preneoplastic PNT2 cells, the androgen-dependent LNCaP and the androgen-independent PC3 and DU145 prostate cancer cell lines. We have studied by specific ELISA assays in conditioned media (CM), the secretion of TGFβ1 and TGFβ2 in basal conditions and after hormonal treatment (DHT or E2) and the expression of TGFβ1 mRNA by Northern blot. We have also compared the effect of fibroblast CM on TGFβ secretion by the different cell types. Compared to PNT2 cells, cancer cell lines secrete lower levels of active TGFβ which are not increased in the presence of fibroblast CM. LNCaP cells respond to androgen or estrogen treatment by a 10-fold increase of active TGFβ secretion while PC3 and DU145 are unresponsive. In conclusion, prostate cancer cell lines have lost part of their ability to secrete and activate TGFβ, and to regulate this secretion through stromal–epithelial interactions. Androgen-sensitive cancer cells may compensate this loss by hormonal regulation.  相似文献   

8.

Background

Understanding drivers for metastasis in human cancer is important for potential development of therapies to treat metastases. The role of loss of TGFβ tumor suppressor activities in the metastatic process is essentially unknown.

Methodology/Principal Findings

Utilizing in vitro and in vivo techniques, we have shown that loss of TGFβ tumor suppressor signaling is necessary to allow the last step of the metastatic process - colonization of the metastatic site. This work demonstrates for the first time that TGFβ receptor reconstitution leads to decreased metastatic colonization. Moreover, we have identified a novel TGFβ/PKA tumor suppressor pathway that acts directly on a known cell survival mechanism that responds to stress with the survivin/XIAP dependent inhibition of caspases that effect apoptosis. The linkage between the TGFβ/PKA transduceome signaling and control of metastasis through induction of cell death was shown by TGFβ receptor restoration with reactivation of the TGFβ/PKA pathway in receptor deficient metastatic colon cancer cells leading to control of aberrant cell survival.

Conclusion/Significance

This work impacts our understanding of the possible mechanisms that are critical to the growth and maintenance of metastases as well as understanding of a novel TGFβ function as a metastatic suppressor. These results raise the possibility that regeneration of attenuated TGFβ signaling would be an effective target in the treatment of metastasis. Our work indicates the clinical potential for developing anti-metastasis therapy based on inhibition of this very important aberrant cell survival mechanism by the multifaceted TGFβ/PKA transduceome induced pathway. Development of effective treatments for metastatic disease is a pressing need since metastases are the major cause of death in solid tumors.  相似文献   

9.
10.
We have previously shown that a transforming factor-β species (TGFβ) is a hormonally regulated negative growth factor in estrogen responsive MCF-7 human breast cancer cells. We now demonstrate that androgen withdrawal leads to a significant stimulation of TGFβ-2 mRNA in the androgen-responsive human prostate carcinoma cell line LNCaP. These data indicate that TGFβ-2 is a marker of (anti)androgen action in human prostate cancer in vitro. Based on these results we addressed the question of whether THGβ-2 represented a marker of (anti)androgen action in prostate cancer in vivo: expression of TGFβ mRNA was determined by RNAase protection analysis in normal and malignant prostate tissue obtained from 9 prostate carcinoma patients without endocrine therapy. In parallel, the nuclear dihydrotestosterone (DHT) concentration was measured as an indicator of androgen stimulation in the same tissues. The following results were obtained. Both normal and cancerous tissues show nuclear accumulation of DHT indicating a functional androgen receptor system. TGFβ-2 is equally expressed in both normal and cancerous tissue. Expression of TGFβ-2 and nuclear DHT concentrations are correlated in both benign and malignant tissue. We conclude that TGFβ-2 is a marker of (anti)hormonal action in androgen-dependent tissue.  相似文献   

11.
12.
13.
An in vitro model system is described for studying the problem of loss of steroid sensitivity in breast cancer cells. Growth of cloned oestrogen-sensitive human breast cancer cells in the long-term absence of steroid gives rise to a population of oestrogen-insensitive cells. In ZR-75-1 cells, the effect is clonal but occurs at high frequency suggesting a mechanism affecting a wide proportion of the cell population synchronously. This does not involve any reduction in oestrogen receptor number. Furthermore, there is no coordinated loss of oestrogen-sensitive molecular markers, showing that oestrogen receptors remain not only present but functional. These growth changes are not accompanied by any loss of growth inhibition by antioestrogen. Although steroid deprivation does not result in loss of oestrogen-sensitive markers, this does not hold true for other steroids. There was a reduction in progestin, androgen and glucocorticoid regulation on transfected LTRs. Loss of steroid-sensitive growth was accompanied by changes in response to exogenous growth factors and altered endogenous growth factor mRNA production. Steroid-deprived T-47-D cells acquire sensitivity to stimulation by TGFβ and have raised TGFβ1 and TGFβ2 mRNA levels. ZR-75-1 cells are growth inhibited by TGFβ and have reduced TGFβ1mRNA levels. In MCF-7 cells, increased IGFII mRNA, following transfection, can result in an increased basal cell growth rate in the absence of steroid. These findings are discussed in relation to possible autocrine mechanisms in the loss of steroid sensitivity of breast cancer cells.  相似文献   

14.
15.
Fibrosis is characterized by elevated transforming growth factor β (TGFβ) signaling, resulting in extracellular matrix accumulation and increased PAI-1 (plasminogen activator inhibitor) expression. PAI-1 induces the internalization of urokinase plasminogen activator/receptor and integrin αvβ3 from the cell surface. Since increased αvβ3 expression correlates with increased TGFβ signaling, we hypothesized that aberrant PAI-1-mediated αvβ3 endocytosis could initiate an autocrine loop of TGFβ activity. We found that in PAI-1 knock-out (KO) mouse embryonic fibroblasts), αvβ3 endocytosis was reduced by ∼75%, leaving αvβ3 in enlarged focal adhesions, similar to wild type cells transfected with PAI-1 small interfering RNA. TGFβ signaling was significantly enhanced in PAI-1 KO cells, as demonstrated by a 3-fold increase in SMAD2/3-containing nuclei and a 2.9-fold increase in TGFβ activity that correlated with an increase in αvβ3 and TGFβ receptor II expression. As expected, PAI-1 KO cells had unregulated plasmin activity, which was only partially responsible for TGFβ activation, as evidenced by a mere 25% reduction in TGFβ activity when plasmin was inhibited. Treatment of cells with an αvβ3-specific cyclic RGD peptide (GpenGRGD) led to a more profound (59%) TGFβ inhibition; a nonspecific RGD peptide (GRGDNP) inhibited TGFβ by only 23%. Human primary fibroblasts were used to confirm that PAI-1 inhibition and β3 overexpression led to an increase in TGFβ activity. Consistent with a fibrotic phenotype, PAI-1 KO cells were constitutively myofibroblasts that had a 1.6-fold increase in collagen deposition over wild type cells. These data suggest that PAI-1-mediated regulation of αvβ3 integrin is critical for the control of TGFβ signaling and the prevention of fibrotic disease.Fibrotic disorders can result from environmental toxins, persistent infection, autoimmune disease, or mechanical injury, leading to the hardening and scarring of tissues. In fibrotic diseases, such as liver cirrhosis, renal fibrosis, and idiopathic lung fibrosis, or in pathological wound healing, such as hypertrophic scarring, scleroderma, and Dupuytren disease, the persistence of myofibroblasts contributes to disease progression by overproduction of extracellular matrix (ECM)2 and by excessive contraction (13). A shift in the balance of growth factors and cytokines that promote ECM deposition and proteases that degrade matrix often contributes to fibrotic disease (4, 5). Plasmin, a broad spectrum protease that is generated from plasminogen by uPA, is one of the proteases that degrades matrix and activates growth factors and other proteases (6). Since uPA activity is inhibited by PAI-1, the overexpression of PAI-1 results in matrix accumulation. For this reason, PAI-1 is a key prognostic marker for fibrotic disease. PAI-1 exerts its inhibitory activity on uPA by stimulating the endocytosis of the cell surface uPA·uPAR complex through the low density lipoprotein receptor-related protein (7). Integrin αvβ3 is also internalized with the uPA·uPAR·low density lipoprotein receptor-related protein complex (8). After endocytosis, uPAR and integrins are recycled back to the cell surface for another round of binding (8, 9). uPAR and αvβ3 promote cellular attachment and spreading, since they are receptors for the extracellular matrix molecule, vitronectin (10). Thus, cycling of the complex is thought to stimulate the attachment and detachment that is necessary for cell migration (8). Consequently, a shift in the expression of any of these components (PAI-1/uPA/uPAR/αvβ3) can result in either aggressive migration, as seen in cancer invasion, or a persistent increase in cell adhesion and cell tension, as seen in myofibroblasts in fibrotic tissue.The family of TGFβ growth factors has been intensively studied for their role in fibrotic wound healing. Up-regulation of TGFβ results in amplified and persistent overproduction of molecules, such as integrins and PAI-1 and other protease inhibitors (e.g. TIMPs) (2, 3). Up-regulated integrins continue the cycle of TGFβ signaling by participating in the sustained activation of TGFβ from its latent form. To date, studies have found that various αv integrins participate in the activation of TGFβ (αvβ3, αvβ5, αvβ6, and αvβ8), but the mechanism differs (1115). Integrins can serve as docking proteins to localize proteases that cleave and activate latent TGFβ in the ECM, or they can directly activate latent TGFβ in a protease-independent manner. Recently, it was discovered that latent TGFβ is also activated by mechanical stress generated from an integrin-mediated interaction between myofibroblasts and the ECM, primarily involving αvβ5. The mechanical stress promotes a conformational change that activates the latent TGFβ complex (15). αv integrins also modulate TGFβ signaling through the binding of αvβ3 to TGFβ receptor II (TGFβRII) in the presence of TGFβ. This interaction was shown to promote a dramatic increase in the proliferation of lung fibroblasts and induce invasion of epithelial breast cancer cells (16, 17).Our data establish a role for the PAI-1-mediated control of αvβ3 expression and support a significant role for αvβ3 in TGFβ signaling. Using PAI-1 KO cells, we tested the hypothesis that the absence of PAI-1 would result in the accumulation of αvβ3 on the cell surface, since PAI-1 promotes the endocytosis of uPA·uPAR·αvβ3. PAI-1-mediated endocytosis of β3 was significantly reduced in the PAI-1 KO cells. Correspondingly, we report that β3 accumulated at the cell surface in enlarged β3-containing focal adhesions. Thus, we explored whether the accumulation of αvβ3 on the cell surface had fibrogenic effects even in the absence of profibrotic PAI-1. Our results demonstrate dramatically increased TGFβ activity and an increase in collagen expression in PAI-1 KO cells. Together, these findings suggest that PAI-1 modulates β3 expression and localization and, in turn, TGFβ signaling. Our data reveal that maintaining precise levels of PAI-1 is a key to preventing fibrosis. Understanding the consequence of regulating PAI-1 activity is critical in light of the many clinical therapies currently under development that target PAI-1 (18, 19).  相似文献   

16.
Promyelocytic leukemia-retinoic acid receptor alpha (PML-RARα) expression in acute promyelocytic leukemia (APL) impairs transforming growth factor beta (TGFβ) signaling, leading to cell growth advantage. Halofuginone (HF), a low-molecular-weight alkaloid that modulates TGFβ signaling, was used to treat APL cell lines and non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice subjected to transplantation with leukemic cells from human chorionic gonadotrophin-PML-RARα transgenic mice (TG). Cell cycle analysis using incorporated bromodeoxyuridine and 7-amino-actinomycin D showed that, in NB4 and NB4-R2 APL cell lines, HF inhibited cellular proliferation (P<0.001) and induced apoptosis (P = 0.002) after a 24-hour incubation. Addition of TGFβ revealed that NB4 cells were resistant to its growth-suppressive effects and that HF induced these effects in the presence or absence of the cytokine. Cell growth inhibition was associated with up-regulation of TGFβ target genes involved in cell cycle regulation (TGFB, TGFBRI, SMAD3, p15, and p21) and down-regulation of MYC. Additionally, TGFβ protein levels were decreased in leukemic TG animals and HF in vivo could restore TGFβ values to normal. To test the in vivo anti-leukemic activity of HF, we transplanted NOD/SCID mice with TG leukemic cells and treated them with HF for 21 days. HF induced partial hematological remission in the peripheral blood, bone marrow, and spleen. Together, these results suggest that HF has anti-proliferative and anti-leukemic effects by reversing the TGFβ blockade in APL. Since loss of the TGFβ response in leukemic cells may be an important second oncogenic hit, modulation of TGFβ signaling may be of therapeutic interest.  相似文献   

17.
Cancer Cell 22: 571–584Metastasis, the process whereby tumour cells disseminate and colonize distant organs, is the primary cause of cancer mortality. Diverse models have been proposed to explain how tumour cells acquire metastatic competency. Calon et al (2012) now provide insight into the molecular underpinnings of metastasis by describing a key stromal, non-cell autonomous role for Transforming Growth Factor-beta (TGFβ) in promoting the initiation of colonization in otherwise TGFβ-resistant colorectal cancer (CRC) cells.Growing tumour cells are surrounded by stroma, a heterogenous population of cells that includes fibroblasts, endothelial precursors and cells of the immune system (Sethi and Kang, 2011; Valastyan and Weinberg, 2011). This stroma engages in an active dialogue with the tumour cells to create a unique microenvironment that is conducive to the survival and progression of a growing tumour. In late stage tumours, productive metastases arise when the tumour cells leave the primary site to disseminate throughout the body and seed new secondary tumours in distant organs. How tumour cells leave behind their primary microenvironment to establish and successfully colonize secondary sites that might harbour tumour-hostile environments has been the subject of extensive research and speculation. A recent study by Calon et al (2012) provides new insights into this long-standing question with the discovery that Transforming Growth Factor-beta (TGFβ) produced by tumour cells critically promotes colorectal cancer (CRC) cell colonization through its actions on the stroma (Figure 1).Open in a separate windowFigure 1TGFβ acting on stromal cells in the primary tumour promotes metastasis. Colorectal cancer cells (CRCs) are frequently insensitive to TGFβ as a result of mutations in pathway components, including the TGFβ receptors (TBRs) and Smads. At the primary tumour site, CRCs that secrete high level of TGFβ induce expression of IL11 in the cancer-associated fibroblasts (CAFs) found in the adjacent stroma. The CRCs then respond to the IL11 via GP130 and this promotes tumour colonization of secondary sites.The secreted factor, TGFβ has been called the ‘Dr Jekyll and Mr Hyde'' of cancer (Bierie and Moses, 2006) due to paradoxical function as both a tumour suppressor and a tumour promoter. For instance, TGFβ inhibits the proliferation of epithelial cells, an activity that most tumour cells must learn to overcome during cancer progression. However, TGFβ also promotes the metastatic phenotype by enhancing tumour cell migration and promoting epithelial-to-mesenchymal transition (EMT).In human CRC, the majority of tumour cells display constitutive Wnt signalling, typically because of mutations in either the adenomas polyposis gene (APC) or β catenin. However, mutations in TGFβ signalling pathway components, including the cell-surface receptors or the intracellular Smad mediator proteins, also play a prominent role, consistent with a tumour suppressive function of TGFβ. Nevertheless, high levels of TGFβ are found in CRC patients and correlates with poor clinical outcome (Tsushima et al, 2001). This raises the question of how TGFβ might promote poor clinical outcome in cancers that have acquired insensitivity to TGFβ. To explore this, Calon et al (2012) examined TGFβ expression levels in a large cohort of CRC patients and noted a strong positive association between the level of TGFβ expression and the risk of cancer recurrence. Indeed, TGFβ expression level outperformed the American Joint Cancer Committee (AJCC) staging system in predictive power. Consistent with the frequent loss of TGFβ pathway mediators in CRC, staining of tumour sections for active TGFβ signalling showed much higher levels in stromal cells as compared to the epithelial CRC cells. Expression profiling for TGFβ-responsive gene signatures (TBRS) using isolated stromal cell populations corroborated this observation, with high levels of TGFβ signalling evident in all stromal cell types tested, including fibroblasts, endothelial and immune cells. However, analysis of in vivo gene expression patterns revealed that it was the TBRS associated with cancer-associated fibroblasts (CAFs) that was the main predictor of poor outcome after therapy.To provide direct evidence of a connection between stromal TGFβ signalling and disease progression, Calon et al (2012) first conducted an elegant series of in vivo experiments in mice, using several colorectal cell lines that have inactivated TGFβ signalling. Subcutaneous injection of variants of these lines engineered to overexpress TGFβ led to activation of TGFβ signalling in adjacent stroma. Calon et al (2012) next turned their attention to examining whether stromal TGFβ signalling might influence metastasis. Inoculation of the engineered CRC cells in the caecum or spleen enhanced the rate of metastasis to the lung and/or liver that was particularly pronounced within the first 24 h post inoculation and most notably was abolished by administration of LY2157299, a TGFβ receptor-selective inhibitor. Similarly, liver metastasis arising through intra-splenic injection of colon cancer stem cells isolated from a patient with TGFβ receptor mutations was abolished by systemic TGFβ receptor-inhibitor treatment. Thus, high levels of TGFβ act to enhance the colonization capability of CRC cells at the initial phase of metastasis.In breast cancer cells, which retain an intact TGFβ signalling pathway, a cell-autonomous role for secreted TGFβ in mediating organ-specific metastatic colonization has been delineated (Kang et al, 2003; Massague, 2008; Padua et al, 2008). However, since the CRC cells employed by Calon et al (2012) were insensitive to TGFβ, the authors focused on the question of how stromal TGFβ signalling might confer a metastatic phenotype to the tumour cells. They went on to show that the IL11, which is secreted by TGFβ-stimulated CAFs, acting through the GP130/STAT3 pathway in the CRC cells, was required for colonization, most likely by suppressing tumour cell apoptosis (Figure 1). This likely allows the tumour cells to survive in the relatively hostile metastatic environments they encounter on their way to distant sites. Indeed, CRC cells engineered to produce their own IL11 effectively colonized liver, lungs, distant lymph nodes and brain.The process of metastasis is extremely inefficient, so how a cell might undergo the genetic and/or epigenetic changes required to leave the primary site and colonize a different organ with a distinct microenvironment is a critical question. Calon et al (2012) now provide new insights into the process by showing that TGFβ signalling in CAFs directed by the tumour cells feeds back on the cancer cell in the primary site to fuel metastasis. These studies thus provide support for the notion that tumour cells acquire the necessary changes to adapt to a new environment while still residing in the primary tumour site. However, it is important to remember that the activity of TGFβ on immune cells is also a potent mechanism that regulates the tumour microenvironment to promote cancer progression (Yang et al, 2010). Although, the TBRS in CAFs was shown to be the most relevant for recurrence rates, the finding that all stromal cell types displayed TGFβ-induced changes raises additional questions for future investigations that explore the extent of cellular interplay in contributing to the tumour-promoting role of TGFβ. Understanding the ongoing dialogue between tumour cells and their microenvironment continues to yield a rich resource of new interventional targets that limit not only primary tumour growth, but also metastasis, the primary cause of cancer death.  相似文献   

18.
19.
20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号