Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Cancer Cell ; 13(1): 23-35, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18167337

ABSTRACT

Aberrant TGFbeta signaling is common in human cancers and contributes to tumor metastasis. Here, we demonstrate that Gr-1+CD11b+ myeloid cells are recruited into mammary carcinomas with type II TGF beta receptor gene (Tgfbr2) deletion and directly promote tumor metastasis. Gr-1+CD11b+ cells infiltrate into the invasive front of tumor tissues and facilitate tumor cell invasion and metastasis through a process involving metalloproteinase activity. This infiltration of Gr-1+CD11b+ cells also results in increased abundance of TGF beta 1 in tumors with Tgfbr2 deletion. The recruitment of Gr-1+CD11b+ cells into tumors with Tgfbr2 deletion involves two chemokine receptor axes, the SDF-1/CXCR4 and CXCL5/CXCR2 axes. Together, these data indicate that Gr-1+CD11b+ cells contribute to TGFbeta-mediated metastasis through enhancing tumor cell invasion and metastasis.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CD11b Antigen/metabolism , Myeloid Cells/pathology , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Breast Neoplasms/enzymology , Cell Line, Tumor , Female , Gene Deletion , Humans , Matrix Metalloproteinases/biosynthesis , Mice , Models, Biological , Myeloid Cells/enzymology , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism
2.
Proc Natl Acad Sci U S A ; 109(8): 2814-9, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-21576484

ABSTRACT

Bone morphogenetic proteins (BMPs) are members of the TGF-Ɵ superfamily of signaling molecules. BMPs can elicit a wide range of effects in many cell types and have previously been shown to induce growth inhibition in carcinoma cells as well as normal epithelia. Recently, it has been demonstrated that BMP4 and BMP7 are overexpressed in human breast cancers and may have tumor suppressive and promoting effects. We sought to determine whether disruption of the BMP receptor 2 (BMPR2) would alter mammary tumor progression in mice that express the Polyoma middle T antigen. Mice expressing Polyoma middle T antigen under the mouse mammary tumor virus promoter were combined with mice that have doxycycline-inducible expression of a dominant-negative (DN) BMPR2. We did not observe any differences in tumor latency. However, mice expressing the BMPR2-DN had a fivefold increase in lung metastases. We characterized several cell autonomous changes and found that BMPR2-DN-expressing tumor cells had higher rates of proliferation. We also identified unique changes in inflammatory cells and secreted chemokines/cytokines that accompanied BMPR2-DN-expressing tumors. By immunohistochemistry, it was found that BMPR2-DN primary tumors and metastases had an altered reactive stroma, indicating specific changes in the tumor microenvironment. Among the changes we discovered were increased myeloid derived suppressor cells and the chemokine CCL9. BMP was shown to directly regulate CCL9 expression. We conclude that BMPR2 has tumor-suppressive function in mammary epithelia and microenvironment and that disruption can accelerate mammary carcinoma metastases.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/metabolism , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Paracrine Communication , Animals , Antigens, Polyomavirus Transforming/metabolism , Cell Movement , Cell Proliferation , Chemokines/metabolism , Disease Progression , Female , Humans , Inflammation/pathology , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/blood supply , Mammary Tumor Virus, Mouse/metabolism , Mice , Myeloid Cells/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic , Signal Transduction , Tumor Microenvironment
3.
Breast Cancer Res ; 16(5): 425, 2014 Oct 04.
Article in English | MEDLINE | ID: mdl-25280532

ABSTRACT

INTRODUCTION: Transforming growth factor beta (TGFƟ) plays a major role in the regulation of tumor initiation, progression, and metastasis. It is depended on the type II TGFƟ receptor (TƟRII) for signaling. Previously, we have shown that deletion of TƟRII in mammary epithelial of MMTV-PyMT mice results in shortened tumor latency and increased lung metastases. However, active TGFƟ signaling increased the number of circulating tumor cells and metastases in MMTV-Neu mice. In the current study, we describe a newly discovered connection between attenuated TGFƟ signaling and human epidermal growth factor receptor 2 (HER2) signaling in mammary tumor progression. METHODS: All studies were performed on MMTV-Neu mice with and without dominant-negative TƟRII (DNIIR) in mammary epithelium. Mammary tumors were analyzed by flow cytometry, immunohistochemistry, and immunofluorescence staining. The levels of secreted proteins were measured by enzyme-linked immunosorbent assay. Whole-lung mount staining was used to quantitate lung metastasis. The Cancer Genome Atlas (TCGA) datasets were used to determine the relevance of our findings to human breast cancer. RESULTS: Attenuated TGFƟ signaling led to a delay tumor onset, but increased the number of metastases in MMTVNeu/DNIIR mice. The DNIIR tumors were characterized by increased vasculogenesis, vessel leakage, and increased expression of vascular endothelial growth factor (VEGF). During DNIIR tumor progression, both the levels of CXCL1/5 and the number of CD11b+Gr1+ cells and T cells decreased. Analysis of TCGA datasets demonstrated a significant negative correlation between TGFBR2 and VEGF genes expression. Higher VEGFA expression correlated with shorter distant metastasis-free survival only in HER2+ patients with no differences in HER2-, estrogen receptor +/- or progesterone receptor +/- breast cancer patients. CONCLUSION: Our studies provide insights into a novel mechanism by which epithelial TGFƟ signaling modulates the tumor microenvironment, and by which it is involved in lung metastasis in HER2+ breast cancer patients. The effects of pharmacological targeting of the TGFƟ pathway in vivo during tumor progression remain controversial. The targeting of TGFƟ signaling should be a viable option, but because VEGF has a protumorigenic effect on HER2+ tumors, the targeting of this protein could be considered when it is associated with attenuated TGFƟ signaling.


Subject(s)
Lung Neoplasms/metabolism , Mammary Neoplasms, Experimental/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Carcinogenesis/metabolism , Chemokines/metabolism , Female , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Mice, Transgenic , Neovascularization, Pathologic/metabolism , Proto-Oncogene Proteins c-akt/metabolism , T-Lymphocytes/immunology , Vascular Endothelial Growth Factor A/metabolism
4.
Breast Cancer Res ; 14(4): R98, 2012 Jul 02.
Article in English | MEDLINE | ID: mdl-22748014

ABSTRACT

INTRODUCTION: Transforming growth factor beta (TGF-Ɵ) has a dual role during tumor progression, initially as a suppressor and then as a promoter. Epithelial TGF-Ɵ signaling regulates fibroblast recruitment and activation. Concurrently, TGF-Ɵ signaling in stromal fibroblasts suppresses tumorigenesis in adjacent epithelia, while its ablation potentiates tumor formation. Much is known about the contribution of TGF-Ɵ signaling to tumorigenesis, yet the role of TGF-Ɵ in epithelial-stromal migration during tumor progression is poorly understood. We hypothesize that TGF-Ɵ is a critical regulator of tumor-stromal interactions that promote mammary tumor cell migration and invasion. METHODS: Fluorescently labeled murine mammary carcinoma cells, isolated from either MMTV-PyVmT transforming growth factor-beta receptor II knockout (TƟRII KO) or TƟRIIfl/fl control mice, were combined with mammary fibroblasts and xenografted onto the chicken embryo chorioallantoic membrane. These combinatorial xenografts were used as a model to study epithelial-stromal crosstalk. Intravital imaging of migration was monitored ex ovo, and metastasis was investigated in ovo. Epithelial RNA from in ovo tumors was isolated by laser capture microdissection and analyzed to identify gene expression changes in response to TGF-Ɵ signaling loss. RESULTS: Intravital microscopy of xenografts revealed that mammary fibroblasts promoted two migratory phenotypes dependent on epithelial TGF-Ɵ signaling: single cell/strand migration or collective migration. At epithelial-stromal boundaries, single cell/strand migration of TƟRIIfl/fl carcinoma cells was characterized by expression of α-smooth muscle actin and vimentin, while collective migration of TƟRII KO carcinoma cells was identified by E-cadherin+/p120+/Ɵ-catenin+ clusters. TƟRII KO tumors also exhibited a twofold greater metastasis than TƟRIIfl/fl tumors, attributed to enhanced extravasation ability. In TƟRII KO tumor epithelium compared with TƟRIIfl/fl epithelium, Igfbp4 and Tspan13 expression was upregulated while Col1α2, Bmp7, Gng11, Vcan, Tmeff1, and Dsc2 expression was downregulated. Immunoblotting and quantitative PCR analyses on cultured cells validated these targets and correlated Tmeff1 expression with disease progression of TGF-Ɵ-insensitive mammary cancer. CONCLUSION: Fibroblast-stimulated carcinoma cells utilize TGF-Ɵ signaling to drive single cell/strand migration but migrate collectively in the absence of TGF-Ɵ signaling. These migration patterns involve the signaling regulation of several epithelial-to-mesenchymal transition pathways. Our findings concerning TGF-Ɵ signaling in epithelial-stromal interactions are important in identifying migratory mechanisms that can be targeted as recourse for breast cancer treatment.


Subject(s)
Cell Communication , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Stromal Cells/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Intercellular Junctions/metabolism , Mice , Neoplasms/genetics , Phenotype , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , beta Catenin/metabolism
5.
Proteomics ; 10(13): 2458-70, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20405477

ABSTRACT

Transforming growth factor beta (TGF-beta) is a master regulator of autocrine and paracrine signaling pathways between a tumor and its microenvironment. Decreased expression of TGF-beta type II receptor (TbetaRII) in stromal cells is associated with increased tumor metastasis and shorter patient survival. In this study, SILAC quantitative proteomics was used to identify differentially externalized proteins in the conditioned media from the mammary fibroblasts with or without intact TbetaRII. Over 1000 proteins were identified and their relative differential levels were quantified. Immunoassays were used to further validate identification and quantification of the proteomic results. Differential expression was detected for various extracellular proteins, including proteases and their inhibitors, growth factors, cytokines, and extracellular matrix proteins. CXCL10, a cytokine found to be up-regulated in the TbetaRII knockout mammary fibroblasts, is shown to directly stimulate breast tumor cell proliferation and migration. Overall, this study revealed hundreds of specific extracellular protein changes modulated by deletion of TbetaRII in mammary fibroblasts, which may play important roles in the tumor microenvironment. These results warrant further investigation into the effects of inhibiting the TGF-beta signaling pathway in fibroblasts because systemic inhibition of TGF-beta signaling pathways is being considered as a potential cancer therapy.


Subject(s)
Fibroblasts/chemistry , Mammary Glands, Animal/chemistry , Proteome/analysis , Signal Transduction , Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Movement , Cell Proliferation , Fibroblasts/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Proteome/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/metabolism
6.
Mol Cancer Res ; 7(5): 645-53, 2009 May.
Article in English | MEDLINE | ID: mdl-19435821

ABSTRACT

A subset of neoplastic cells within human high-grade gliomas has features associated with stem cells. These cells may sustain glioma growth, and their stem-like properties may confer resistance to standard glioma treatments. Whether glioma stem cells derive from indigenous neural stem cells (NSC), or from tumor cells that have reacquired stem cell-like properties, is unknown. However, signaling pathways that are tightly regulated and central to NSC biology, including the Ras/Raf/Erk pathway, are hyperactive and pathogenic in gliomagenesis. Furthermore, data in animal models suggests that, in some cases, tumors are initiated in the subventricular zone (SVZ), a stem/progenitor cell niche in the mature brain. We activated oncogenic K-ras in mouse glioneuronal precursor cells and adult SVZ cells using GFAP-Cre. GFAP-Cre+/K-ras(G12D) mice showed a marked expansion of glial fibriallary acidic protein (GFAP)- and TUJ1-expressing cell populations in the SVZ. In addition, mice developed intermediate grade, infiltrating glioma with 100% penetrance. Tumors were consistently located in the amygdalohippocampal region and nearby cortex, often in association with the lateral ventricle and expanded SVZ. Tumor cells expressed markers associated with neural progenitor cells, including Olig2, Bmi-1, and PDGFR-alpha. These data suggest that infiltrating tumor cells may arise from NSC transformed by activation of oncogenic K-ras in vivo.


Subject(s)
Cerebral Ventricles/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glioma/metabolism , Integrases/metabolism , ras Proteins/metabolism , Animals , Cerebral Ventricles/pathology , Female , Glial Fibrillary Acidic Protein/genetics , Glioma/pathology , Immunohistochemistry , Integrases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neurons/metabolism , Neurons/pathology , ras Proteins/genetics
7.
Cancer Biol Ther ; 6(4): 561-70, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17495520

ABSTRACT

Breast cancers often have deregulated hepatocyte growth factor (HGF) and c-Met signaling that results in increased tumor growth and invasion. Elucidating the mechanism responsible for HGF/c-Met action in breast cancer progression has been difficult as c-Met communicates with a number of secondary receptors that can lead to various pathological outcomes. Understanding how these secondary receptors facilitate HGF/c-Met cellular responses will aid in the development of better therapeutic treatment options for breast cancer patients with elevated HGF signaling. In the present study it was shown that the epidermal growth factor receptor (EGFR) plays a significant role in HGF/c-Met mediated biological activities indicative of advanced tumor pathology, including enhanced proliferation and invasion. The clinically relevant EGFR inhibitor gefitinib was used to determine the role of EGFR in HGF-induced proliferation and motility in several mammary carcinoma cells including PyVmT, MDA-MB-231 and 4T1. Our analyses indicated that EGFR inhibition significantly blocked HGF activation of c-Met and EGFR and that inhibition of these pathways mitigated HGF induced proliferation and motility. The data indicate that this inhibition was not through a direct effect of gefitinib on c-Met, but that EGFR is necessary for c-Met activation in the assays performed. These results provide a novel mechanism of action for EGFR as a mediator of HGF signaling thereby linking EGFR to the oncogenic potential of c-Met in mammary carcinomas cells.


Subject(s)
Breast Neoplasms/enzymology , Carcinoma/enzymology , Drug Resistance, Neoplasm , ErbB Receptors/physiology , Hepatocyte Growth Factor/metabolism , Mammary Glands, Human/enzymology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/enzymology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Female , Gefitinib , Hepatocyte Growth Factor/pharmacology , Humans , Mammary Glands, Human/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , Receptor Protein-Tyrosine Kinases/metabolism
8.
Mol Cell Biol ; 22(23): 8184-98, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12417722

ABSTRACT

Transforming growth factor beta (TGF-beta) induces cell cycle arrest of most nontransformed epithelial cell lines. In contrast, many human carcinomas are refractory to the growth-inhibitory effect of TGF-beta. TGF-beta overexpression inhibits tumorigenesis, and abolition of TGF-beta signaling accelerates tumorigenesis, suggesting that TGF-beta acts as a tumor suppressor in mouse models of cancer. A screen to identify agents that potentiate TGF-beta-induced growth arrest demonstrated that the potential anticancer agent rapamycin cooperated with TGF-beta to induce growth arrest in multiple cell lines. Rapamycin also augmented the ability of TGF-beta to inhibit the proliferation of E2F1-, c-Myc-, and (V12)H-Ras-transformed cells, even though these cells were insensitive to TGF-beta-mediated growth arrest in the absence of rapamycin. Rapamycin potentiation of TGF-beta-induced growth arrest could not be explained by increases in TGF-beta receptor levels or rapamycin-induced dissociation of FKBP12 from the TGF-beta type I receptor. Significantly, TGF-beta and rapamycin cooperated to induce growth inhibition of human carcinoma cells that are resistant to TGF-beta-induced growth arrest, and arrest correlated with a suppression of Cdk2 kinase activity. Inhibition of Cdk2 activity was associated with increased binding of p21 and p27 to Cdk2 and decreased phosphorylation of Cdk2 on Thr(160). Increased p21 and p27 binding to Cdk2 was accompanied by decreased p130, p107, and E2F4 binding to Cdk2. Together, these results indicate that rapamycin and TGF-beta cooperate to inhibit the proliferation of nontransformed cells and cancer cells by acting in concert to inhibit Cdk2 activity.


Subject(s)
Antibiotics, Antineoplastic/metabolism , CDC2-CDC28 Kinases , Carcinoma/metabolism , Cell Division/physiology , Proteins , Sirolimus/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Line , Cell Transformation, Neoplastic , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , DNA-Binding Proteins/metabolism , E2F4 Transcription Factor , Enzyme Inhibitors/metabolism , Epithelial Cells/physiology , Genes, Reporter , Growth Inhibitors/metabolism , Humans , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , Signal Transduction/physiology , Tacrolimus Binding Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
9.
Cancer Res ; 65(6): 2296-302, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15781643

ABSTRACT

Transforming growth factor-beta (TGF-beta) isoforms are growth factors that function physiologically to regulate development, cellular proliferation, and immune responses. The role of TGF-beta signaling in mammary tumorigenesis is complex, as TGF-beta has been reported to function as both a tumor suppressor and tumor promoter. To elucidate the role of TGF-beta signaling in mammary gland development, tumorigenesis, and metastasis, the gene encoding type II TGF-beta receptor, Tgfbr2, was conditionally deleted in the mammary epithelium (Tgfbr2MGKO). Loss of Tgfbr2 in the mammary epithelium results in lobular-alveolar hyperplasia in the developing mammary gland and increased apoptosis. Tgfbr2MGKO mice were mated to the mouse mammary tumor virus-polyomavirus middle T antigen (PyVmT) transgenic mouse model of metastatic breast cancer. Loss of Tgfbr2 in the context of PyVmT expression results in a shortened median tumor latency and an increased formation of pulmonary metastases. Thus, our studies support a tumor-suppressive role for epithelial TGF-beta signaling in mammary gland tumorigenesis and show that pulmonary metastases can occur and are even enhanced in the absence of TGF-beta signaling in the carcinoma cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , Mammary Glands, Animal/physiology , Mammary Neoplasms, Experimental/genetics , Receptors, Transforming Growth Factor beta/genetics , Animals , Antigens, Polyomavirus Transforming/physiology , Cell Growth Processes/physiology , Cell Transformation, Neoplastic/pathology , Epithelial Cells/cytology , Epithelial Cells/physiology , Female , Hyperplasia , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Male , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases , Pulmonary Alveoli/pathology , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/deficiency , Transgenes/genetics
10.
Mol Cancer Ther ; 1(10): 759-68, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12492108

ABSTRACT

Transforming growth factor beta (TGF-beta) is a multifunctional protein that has been shown to possess potent growth-inhibitory activity. To identify small molecular weight compounds with TGF-beta-like activities, high throughput screening was performed using mink lung epithelial cells stably transfected with a TGF-beta-responsive plasminogen activator inhibitor 1 promoter/luciferase construct. Biaryl hydroxamate compounds were identified that demonstrated TGF-beta-like activities. 7-[4-(4-cyanophenyl)phenoxy]-heptanohydroxamic acid (A-161906) demonstrated complete TGF-beta-like agonist activity in the plasminogen activator inhibitor 1/luciferase construct. A-161906 inhibited the proliferation of multiple cell lines in a concentration-dependent manner. Cells were growth arrested at the G1-S checkpoint similar to TGF-beta. Consistent with the G1-S arrest, A-161906 induced the expression of the cyclin-dependent kinase inhibitor p21waf1/cip1. A-161906 produced many cellular effects similar to that of TGF-beta but did not displace labeled TGF-beta from its receptors. Cells with mutations in either of the TGF-beta receptors I or II were growth-arrested by A-161906. Therefore, the site of action of A-161906 appears to be distal to the receptors and possibly involved with the signaling events controlled by TGF-beta. The TGF-beta mimetic effect of A-161906 can be partially, if not entirely, explained by its activity as a histone deacetylase (HDAC) inhibitor. A-161906 demonstrated potent HDAC-inhibitory activity (IC50 = 9 nM). A-161906 is a novel small molecular weight compound (< 400 MW) having TGF-beta mimetic activity as a result of its potent HDAC-inhibitory activity. These results and those of others demonstrate the importance of HDACs in regulation of the TGF-beta signaling pathway(s).


Subject(s)
Biphenyl Compounds/pharmacology , Hydroxamic Acids/pharmacology , Transforming Growth Factor beta/pharmacology , Acetylation , Animals , Blotting, Western , Cell Cycle , Cell Division , Cell Line , Collagenases/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Epithelial Cells/metabolism , Fibroblasts/metabolism , Fibronectins/biosynthesis , G1 Phase , Gelsolin/metabolism , Histone Deacetylase Inhibitors , Humans , Inhibitory Concentration 50 , Keratinocytes/metabolism , Luciferases/metabolism , Lung/cytology , Mice , Mink , Models, Chemical , Phenotype , Plasminogen Activator Inhibitor 1/genetics , Promoter Regions, Genetic , S Phase , Time Factors , Transfection , Transforming Growth Factor beta/chemistry
11.
PLoS One ; 10(1): e0117908, 2015.
Article in English | MEDLINE | ID: mdl-25629162

ABSTRACT

Metastasis is the most devastating aspect of cancer, however we know very little about the mechanisms of local invasion, the earliest step of metastasis. During tumor growth CD11b+ Gr1+ cells, known also as MDSCs, have been shown to promote tumor progression by a wide spectrum of effects that suppress the anti-tumor immune response. In addition to immunosuppression, CD11b+ Gr1+ cells promote metastasis by mechanisms that are currently unknown. CD11b+ Gr1+ cells localize near fibroblasts, which remodel the ECM and leave tracks for collective cell migration of carcinoma cells. In this study we discovered that CD11b+ Gr1+ cells promote invasion of mammary carcinoma cells by increasing fibroblast migration. This effect was directed by secreted factors derived from CD11b+ Gr1+ cells. We have identified several CD11b+ Gr1+ cell secreted proteins that activate fibroblast migration, including CXCL11, CXCL15, FGF2, IGF-I, IL1Ra, Resistin, and Shh. The combination of CXCL11 and FGF2 had the strongest effect on fibroblast migration that is associated with Akt1 and ERK1/2 phosphorylation. Analysis of subsets of CD11b+ Gr1+ cells identified that CD11b+ Ly6Chigh Ly6Glow cells increase fibroblast migration more than other myeloid cell populations. Additionally, tumor-derived CD11b+ Gr1+ cells promote fibroblast migration more than splenic CD11b+ Gr1+ cells of tumor-bearing mice. While TGFƟ signaling in fibroblasts does not regulate their migration toward CD11b+ Gr1+ cells, however deletion of TGFƟ receptor II on CD11b+ Gr1+ cells downregulates CXCL11, Shh, IGF1 and FGF2 resulting in reduced fibroblast migration. These studies show that TGFƟ signaling in CD11b+ Gr1+ cells promotes fibroblast directed carcinoma invasion and suggests that perivascular CD11b+ Ly6Chigh Ly6Glow cells may be the stimulus for localized invasion leading to metastasis.


Subject(s)
Fibroblasts/metabolism , Mammary Neoplasms, Experimental/metabolism , Myeloid Cells/metabolism , Neoplasm Invasiveness/pathology , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Line, Tumor , Cell Movement/physiology , Female , Fibroblasts/pathology , Mammary Neoplasms, Experimental/pathology , Mice , Myeloid Cells/pathology , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction/physiology
12.
Neoplasia ; 6(5): 603-10, 2004.
Article in English | MEDLINE | ID: mdl-15548370

ABSTRACT

Transforming growth factor-beta1 (TGF-beta1) can be tumor-suppressive through the activation of the Smad-mediated signaling pathway. TGF-beta1 can also enhance tumor progression by stimulating epithelial-to-mesenchymal transition (EMT) through additional pathways. EMT is characterized by the acquisition of a fibroblast-like cell morphology, dissolution of tight junctions, disruption of adherence junctions, and formation of actin stress fibers. There is evidence linking the activation of mitogen-activated protein kinase pathways to the induction of TGF-beta1-mediated EMT. However, the role of Erk in the induction of TGF-beta1-mediated EMT remains unclear. TGF-beta1 treatment of normal murine mammary gland (NMuMG) epithelial cells resulted in increased gene expression of Ras, Raf, MEK1/2, and Erk1/2, as shown by microarray analysis and real-time polymerase chain reaction. Upon 24 and 48 hours of treatment with TGF-beta1, NMuMG and mouse cortical tubule (MCT) epithelial cells underwent EMT as shown by changes in cell morphology, delocalization of zonula occludens-1 and E-cadherin from cell-cell junctions, and formation of actin stress fibers. TGF-beta1 treatment also resulted in increased levels of phosphorylated Erk and Erk kinase activity. Treatment with an MEK inhibitor, U0126, inhibited increased Erk phosphorylation and kinase activity, and blocked TGF-beta1-induced EMT in both cell lines. These data show that TGF-beta1 induces the activation of the Erk signaling pathway, which is required for TGF-beta1-mediated EMT in vitro.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/enzymology , MAP Kinase Signaling System , Transforming Growth Factor beta/physiology , Animals , Butadienes/pharmacology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Epithelial Cells/drug effects , Gene Expression/drug effects , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/physiology , Mammary Glands, Animal/cytology , Mesoderm/cytology , Mice , Nitriles/pharmacology , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1 , Up-Regulation
13.
Breast Cancer Res ; 5(6): R187-98, 2003.
Article in English | MEDLINE | ID: mdl-14580254

ABSTRACT

BACKGROUND: Transforming growth factor beta (TGF-beta) plays an essential role in a wide array of cellular processes. The most well studied TGF-beta response in normal epithelial cells is growth inhibition. In some cell types, TGF-beta induces an epithelial to mesenchymal transition (EMT). NMuMG is a nontransformed mouse mammary gland epithelial cell line that exhibits both a growth inhibitory response and an EMT response to TGF-beta, rendering NMuMG cells a good model system for studying these TGF-beta effects. METHOD: A National Institutes of Aging mouse 15,000 cDNA microarray was used to profile the gene expression of NMuMG cells treated with TGF-beta1 for 1, 6, or 24 hours. Data analyses were performed using GenePixPro and GeneSpring software. Selected microarray results were verified by northern analyses. RESULTS: Of the 15,000 genes examined by microarray, 939 were upregulated or downregulated by TGF-beta. This represents approximately 10% of the genes examined, minus redundancy. Seven genes previously not known to be regulated by TGF-beta at the transcriptional level (Akt and RhoB) or not at all (IQGAP1, mCalpain, actinin alpha3, Ikki, PP2A-PR53), were identified and their regulation by TGF-beta verified by northern blotting. Cell cycle pathway examination demonstrated downregulation of cyclin D2, c-myc, Id2, p107, E2F5, cyclin A, cyclin B, and cyclin H. Examination of cell adhesion-related genes revealed upregulation of c-Jun, alpha-actinin, actin, myosin light chain, p120cas catenin (Catns), alpha-integrin, integrin beta5, fibronectin, IQGAP1, and mCalpain. CONCLUSION: Using a cDNA microarray to examine TGF-beta-regulated gene expression in NMuMG cells, we have shown regulation of multiple genes that play important roles in cell cycle control and EMT. In addition, we have identified several novel TGF-beta-regulated genes that may mediate previously unknown TGF-beta functions.


Subject(s)
Epithelial Cells/drug effects , Mammary Glands, Animal/drug effects , Transforming Growth Factor beta/pharmacology , Animals , Cell Adhesion/genetics , Cell Cycle/genetics , Cell Line , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Models, Biological , Oligonucleotide Array Sequence Analysis
14.
Breast Cancer Res ; 6(3): R215-31, 2004.
Article in English | MEDLINE | ID: mdl-15084245

ABSTRACT

INTRODUCTION: Transforming growth factor (TGF)-beta1 is proposed to inhibit the growth of epithelial cells in early tumorigenesis, and to promote tumor cell motility and invasion in the later stages of carcinogenesis through the induction of an epithelial to mesenchymal transition (EMT). EMT is a multistep process that is characterized by changes in cell morphology and dissociation of cell-cell contacts. Although there is growing interest in TGF-beta1-mediated EMT, the phenotype is limited to only a few murine cell lines and mouse models. METHODS: To identify alternative cell systems in which to study TGF-beta1-induced EMT, 18 human and mouse established cell lines and cultures of two human primary epithelial cell types were screened for TGF-beta1-induced EMT by analysis of cell morphology, and localization of zonula occludens-1, E-cadherin, and F-actin. Sensitivity to TGF-beta1 was also determined by [3H]thymidine incorporation, flow cytometry, phosphorylation of Smad2, and total levels of Smad2 and Smad3 in these cell lines and in six additional cancer cell lines. RESULTS: TGF-beta1 inhibited the growth of most nontransformed cells screened, but many of the cancer cell lines were insensitive to the growth inhibitory effects of TGF-beta1. In contrast, TGF-beta1 induced Smad2 phosphorylation in the majority of cell lines, including cell lines resistant to TGF-beta1-mediated cell cycle arrest. Of the cell lines screened only two underwent TGF-beta1-induced EMT. CONCLUSION: The results presented herein show that, although many cancer cell lines have lost sensitivity to the growth inhibitory effect of TGF-beta1, most show evidence of TGF-beta1 signal transduction, but only a few cell lines undergo TGF-beta1-mediated EMT.


Subject(s)
Epithelial Cells/cytology , Neoplasm Invasiveness/physiopathology , Transforming Growth Factor beta/physiology , Actins/analysis , Animals , Cadherins/analysis , Cell Adhesion/physiology , Cell Cycle/physiology , Cell Line, Transformed/cytology , Cell Line, Tumor/cytology , Cell Movement/physiology , Cells, Cultured/cytology , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , Disease Progression , Epithelial Cells/metabolism , Humans , In Vitro Techniques , Membrane Proteins/analysis , Mesoderm , Mice , Neoplasm Proteins/physiology , Phenotype , Phosphoproteins/analysis , Phosphorylation , Protein Processing, Post-Translational , Smad2 Protein , Smad3 Protein , Trans-Activators/analysis , Trans-Activators/metabolism , Transforming Growth Factor beta1 , Zonula Occludens-1 Protein
15.
Cancer Res ; 73(17): 5336-46, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23856251

ABSTRACT

The tumor stromal environment can dictate many aspects of tumor progression. A complete understanding of factors driving stromal activation and their role in tumor metastasis is critical to furthering research with the goal of therapeutic intervention. Polyoma middle T-induced mammary carcinomas lacking the type II TGF-Ɵ receptor (PyMT(mgko)) are highly metastatic compared with control PyMT-induced carcinomas (PyMT(fl/fl)). We hypothesized that the PyMT(mgko)-activated stroma interacts with carcinoma cells to promote invasion and metastasis. We show that the extracellular matrix associated with PyMT(mgko) tumors is stiffer and has more fibrillar collagen and increased expression of the collagen crosslinking enzyme lysyl oxidase (LOX) compared with PyMT(fl/fl) mammary carcinomas. Inhibition of LOX activity in PyMT(mgko) mice had no effect on tumor latency and size, but significantly decreased tumor metastasis through inhibition of tumor cell intravasation. This phenotype was associated with a decrease in keratin 14-positive myoepithelial cells in PyMT(mgko) tumors following LOX inhibition as well as a decrease in focal adhesion formation. Interestingly, the primary source of LOX was found to be activated fibroblasts. LOX expression in these fibroblasts can be driven by myeloid cell-derived TGF-Ɵ, which is significantly linked to human breast cancer. Overall, stromal expansion in PyMT(mgko) tumors is likely caused through the modulation of immune cell infiltrates to promote fibroblast activation. This feeds back to the epithelium to promote metastasis by modulating phenotypic characteristics of basal cells. Our data indicate that epithelial induction of microenvironmental changes can play a significant role in tumorigenesis and attenuating these changes can inhibit metastasis. Cancer Res; 73(17); 5336-46. Ā©2013 AACR.


Subject(s)
Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Protein Serine-Threonine Kinases/physiology , Protein-Lysine 6-Oxidase/metabolism , Receptors, Transforming Growth Factor beta/physiology , Stromal Cells/enzymology , Transforming Growth Factor beta/physiology , Animals , Carcinogenesis , Collagen/metabolism , Enzyme Inhibitors/pharmacology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Focal Adhesion Kinase 1/metabolism , Humans , In Situ Hybridization , Keratin-14/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Transgenic , Microscopy, Atomic Force , Myeloid Cells/metabolism , Myeloid Cells/pathology , Phosphorylation , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type II , Signal Transduction , Stromal Cells/pathology
16.
Cancer Discov ; 1(5): 430-41, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22408746

ABSTRACT

We report that IL-17 significantly increases the secretion of CXCL1 and CXCL5 from mammary carcinoma cells, which is downregulated by TGF-Ɵ through the type II TGF-Ɵ receptor (TƟRII). Carcinoma cells with conditional knockout of TƟRII (Tgfbr2(KO)) have enhanced sensitivity to IL-17a in the stimulation of chemokine secretion. During polyoma middle T (PyMT) induced tumor progression, levels of Th17 inducing cytokines TGF-Ɵ, IL-6, IL-23 were increased in PyMT/Tgfbr2(KO) tumors, which was associated with an increased number of Th17 cells. IL-17 increased the suppressive function of MDSCs on T cells through the upregulation of Arg, IDO, and COX2. Treatment of PyMT/Tgfbr2(KO) mice with anti-IL-17 Ab decreased carcinoma growth and metastatic burden. Analysis of human breast cancer transcriptome databases showed a strong association between IL-17 gene expression and poor outcome in lymph node positive, estrogen receptor negative or luminal B subtypes suggesting potential therapeutic approaches.


Subject(s)
Breast Neoplasms/metabolism , Interleukin-17/metabolism , Mammary Neoplasms, Experimental/metabolism , Protein Serine-Threonine Kinases/deficiency , Receptors, Transforming Growth Factor beta/deficiency , Th17 Cells/metabolism , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Interleukin-17/genetics , Ligands , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Neoplasm Metastasis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transcriptome , alpha-Fetoproteins/genetics , alpha-Fetoproteins/metabolism
17.
J Clin Invest ; 121(10): 4106-17, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21926469

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC), one of the most lethal neoplasms, is characterized by an expanded stroma with marked fibrosis (desmoplasia). We previously generated pancreas epithelium-specific TGF-Ɵ receptor type II (Tgfbr2) knockout mice in the context of Kras activation (mice referred to herein as Kras+Tgfbr2KO mice) and found that they developed aggressive PDAC that recapitulated the histological manifestations of the human disease. The mouse PDAC tissue showed strong expression of connective tissue growth factor (Ctgf), a profibrotic and tumor-promoting factor, especially in the tumor-stromal border area, suggesting an active tumor-stromal interaction. Here we show that the PDAC cells in Kras+Tgfbr2KO mice secreted much higher levels of several Cxc chemokines compared with mouse pancreatic intraepithelial neoplasia cells, which are preinvasive. The Cxc chemokines induced Ctgf expression in the pancreatic stromal fibroblasts, not in the PDAC cells themselves. Subcutaneous grafting studies revealed that the fibroblasts enhanced growth of PDAC cell allografts, which was attenuated by Cxcr2 inhibition. Moreover, treating the Kras+Tgfbr2KO mice with the CXCR2 inhibitor reduced tumor progression. The decreased tumor progression correlated with reduced Ctgf expression and angiogenesis and increased overall survival. Taken together, our data indicate that tumor-stromal interactions via a Cxcr2-dependent chemokine and Ctgf axis can regulate PDAC progression. Further, our results suggest that inhibiting tumor-stromal interactions might be a promising therapeutic strategy for PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Pancreatic Neoplasms/therapy , Receptors, Interleukin-8B/antagonists & inhibitors , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/physiopathology , Chemokines, CXC/physiology , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/physiology , Female , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/physiopathology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/physiology , Receptor, Transforming Growth Factor-beta Type II , Receptors, Interleukin-8B/physiology , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Stromal Cells/pathology , Stromal Cells/physiology , Tumor Microenvironment/physiology
18.
J Clin Invest ; 119(6): 1571-82, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19451693

ABSTRACT

In human breast cancer, loss of carcinoma cell-specific response to TGF-beta signaling has been linked to poor patient prognosis. However, the mechanisms through which TGF-beta regulates these processes remain largely unknown. In an effort to address this issue, we have now identified gene expression signatures associated with the TGF-beta signaling pathway in human mammary carcinoma cells. The results strongly suggest that TGF-beta signaling mediates intrinsic, stromal-epithelial, and host-tumor interactions during breast cancer progression, at least in part, by regulating basal and oncostatin M-induced CXCL1, CXCL5, and CCL20 chemokine expression. To determine the clinical relevance of our results, we queried our TGF-beta-associated gene expression signatures in 4 human breast cancer data sets containing a total of 1,319 gene expression profiles and associated clinical outcome data. The signature representing complete abrogation of TGF-beta signaling correlated with reduced relapse-free survival in all patients; however, the strongest association was observed in patients with estrogen receptor-positive (ER-positive) tumors, specifically within the luminal A subtype. Together, the results suggest that assessment of TGF-beta signaling pathway status may further stratify the prognosis of ER-positive patients and provide novel therapeutic approaches in the management of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemokines/biosynthesis , Signal Transduction/drug effects , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Antigens, Viral, Tumor/immunology , Breast Neoplasms/genetics , Cell Differentiation , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Polyomavirus/immunology , Prognosis , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Stem Cells/cytology , Stem Cells/metabolism
19.
Cancer Res ; 68(6): 1809-19, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18339861

ABSTRACT

Transforming growth factor (TGF)-beta signaling has been associated with early tumor suppression and late tumor progression; however, many of the mechanisms that mediate these processes are not known. Using Cre/LoxP technology, with the whey acidic protein promoter driving transgenic expression of Cre recombinase (WAP-Cre), we have now ablated the type II TGF-beta receptor (T beta RII) expression specifically within mouse mammary alveolar progenitors. Transgenic expression of the polyoma virus middle T antigen, under control of the mouse mammary tumor virus enhancer/promoter, was used to produce mammary tumors in the absence or presence of Cre (T beta RII((fl/fl);PY) and T beta RII((fl/fl);PY;WC), respectively). The loss of TGF-beta signaling significantly decreased tumor latency and increased the rate of pulmonary metastasis. The loss of TGF-beta signaling was significantly correlated with increased tumor size and enhanced carcinoma cell survival. In addition, we observed significant differences in stromal fibrovascular abundance and composition accompanied by increased recruitment of F4/80(+) cell populations in T beta RII((fl/fl);PY;WC) mice when compared with T beta RII((fl/fl);PY) controls. The recruitment of F4/80(+) cells correlated with increased expression of known inflammatory genes including Cxcl1, Cxcl5, and Ptgs2 (cyclooxygenase-2). Notably, we also identified an enriched K5(+) dNp63(+) cell population in primary T beta RII((fl/fl);PY;WC) tumors and corresponding pulmonary metastases, suggesting that loss of TGF-beta signaling in this subset of carcinoma cells can contribute to metastasis. Together, our current results indicate that loss of TGF-beta signaling in mammary alveolar progenitors may affect tumor initiation, progression, and metastasis through regulation of both intrinsic cell signaling and adjacent stromal-epithelial interactions in vivo.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Transforming Growth Factor beta/metabolism , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Breast Cyst/metabolism , Breast Cyst/pathology , Cell Differentiation/physiology , Cell Survival/physiology , Disease Progression , Hyperplasia/metabolism , Hyperplasia/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Stromal Cells/metabolism , Stromal Cells/pathology
20.
Genes Dev ; 20(22): 3147-60, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17114585

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an almost uniformly lethal disease in humans. Transforming growth factor-beta (TGF-beta) signaling plays an important role in PDAC progression, as indicated by the fact that Smad4, which encodes a central signal mediator downstream from TGF-beta, is deleted or mutated in 55% and the type II TGF-beta receptor (Tgfbr2) gene is altered in a smaller subset of human PDAC. Pancreas-specific Tgfbr2 knockout mice have been generated, alone or in the context of active Kras (Kras(G12D)) expression, using the Cre-loxP system driven by the endogenous Ptf1a (pancreatic transcription factor-1a) locus. Pancreas-selective Tgfbr2 knockout alone gave no discernable phenotype in 1.5 yr. Pancreas-specific Kras(G12D) activation alone essentially generated only intraepithelial neoplasia within 1 yr. In contrast, the Tgfbr2 knockout combined with Kras(G12D) expression developed well-differentiated PDAC with 100% penetrance and a median survival of 59 d. Heterozygous deletion of Tgfbr2 with Kras(G12D) expression also developed PDAC, which indicated a haploinsufficiency of TGF-beta signaling in this genetic context. The clinical and histopathological manifestations of the combined Kras(G12D) expression and Tgfbr2 knockout mice recapitulated human PDAC. The data show that blockade of TGF-beta signaling and activated Ras signaling cooperate to promote PDAC progression.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Gene Expression , Genes, ras , Pancreatic Neoplasms/pathology , Signal Transduction , Transforming Growth Factor beta/metabolism , Aging , Animals , Connective Tissue Growth Factor , Disease Progression , Epithelium/pathology , Heterozygote , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Neoplasm Invasiveness , Pancreas/cytology , Pancreas/growth & development , Pancreas/pathology , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/deficiency , Stromal Cells/cytology , Stromal Cells/pathology , Transcription Factors/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL