Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Oncotarget ; 7(25): 38164-38179, 2016 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-27203217

RESUMO

Transforming growth factor-ßs (TGF-ßs) regulate tissue homeostasis, and their expression is perturbed in many diseases. The three isoforms (TGF-ß1, -ß2, and -ß3) have similar bioactivities in vitro but show distinct activities in vivo. Little quantitative information exists for expression of TGF-ß isoform proteins in physiology or disease. We developed an optimized method to quantitate protein levels of the three isoforms, using a Luminex® xMAP®-based multianalyte assay following acid-ethanol extraction of tissues. Analysis of multiple tissues and plasma from four strains of adult mice showed that TGF-ß1 is the predominant isoform with TGF-ß2 being ~10-fold lower. There were no sex-specific differences in isoform expression, but some tissues showed inter-strain variation, particularly for TGF-ß2. The only adult tissue expressing appreciable TGF-ß3 was the mammary gland, where its levels were comparable to TGF-ß1. In situ hybridization showed the luminal epithelium as the major source of all TGF-ß isoforms in the normal mammary gland. TGF-ß1 protein was 3-8-fold higher in three murine mammary tumor models than in normal mammary gland, while TGF-ß3 protein was 2-3-fold lower in tumors than normal tissue, suggesting reciprocal regulation of these isoforms in mammary tumorigenesis.


Assuntos
Neoplasias Mamárias Experimentais/imunologia , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Feminino , Humanos , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Isoformas de Proteínas
2.
PLoS One ; 10(5): e0126483, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25993439

RESUMO

Transforming growth factor-ß (TGF-ß) has a dual role in tumorigenesis, acting as either a tumor suppressor or as a pro-oncogenic factor in a context-dependent manner. Although TGF-ß antagonists have been proposed as anti-metastatic therapies for patients with advanced stage cancer, how TGF-ß mediates metastasis-promoting effects is poorly understood. Establishment of TGF-ß-related protein expression signatures at the metastatic site could provide new mechanistic information and potentially allow identification of novel biomarkers for clinical intervention to discriminate TGF-ß oncogenic effects from tumor suppressive effects. In the present study, we found that systemic administration of the TGF-ß receptor kinase inhibitor, SB-431542, significantly inhibited lung metastasis from transplanted 4T1 mammary tumors in Balb/c mice. The differentially expressed proteins in the comparison of lung metastases from SB-431542 treated and control vehicle-treated groups were analyzed by a quantitative LTQ Orbitrap Velos system coupled with stable isotope dimethyl labeling. A total of 36,239 peptides from 6,694 proteins were identified, out of which 4,531 proteins were characterized as differentially expressed. A subset of upregulated proteins in the control group was validated by western blotting and immunohistochemistry. The eukaryotic initiation factor (eIF) family members constituted the most enriched protein pathway in vehicle-treated compared with SB-43512-treated lung metastases, suggesting that increased protein expression of specific eIF family members, especially eIF4A1 and eEF2, is related to the metastatic phenotype of advanced breast cancer and can be down-regulated by TGF-ß pathway inhibitors. Thus our proteomic approach identified eIF pathway proteins as novel potential mediators of TGF-ß tumor-promoting activity.


Assuntos
Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/metabolismo , Proteínas de Neoplasias/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Benzamidas/farmacologia , Linhagem Celular Tumoral , Dioxóis/farmacologia , Fatores de Iniciação em Eucariotos/metabolismo , Feminino , Humanos , Neoplasias Pulmonares/prevenção & controle , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Proteoma/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/antagonistas & inibidores
3.
Biomicrofluidics ; 9(2): 022402, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25825613

RESUMO

This research presents a multiple enzyme-doped thread-based microfluidic system for blood urea nitrogen (BUN) and glucose detection in human whole blood. A novel enzyme-doped thread coated with a thin polyvinylchloride (PVC) membrane is produced for on-site electrochemical detection of urea and glucose in whole blood. Multiple enzymes can be directly applied to the thread without delicate pretreatment or a surface modification process prior to sealing the thread with PVC membrane. Results indicate that the developed device exhibits a good linear dynamic range for detecting urea and glucose in concentrations from 0.1 mM-10.0 mM (R(2 )= 0.9850) and 0.1 mM-13.0 mM (R(2 )= 0.9668), which is suitable for adoption in detecting the concentrations of blood urea nitrogen (BUN, 1.78-7.12 mM) and glucose (3.89-6.11 mM) in serum. The detection result also shows that the developed thread-based microfluidic system can successfully separate and detect the ions, BUN, and glucose in blood. The calculated concentrations of BUN and glucose ante cibum (glucose before meal) in the whole blood sample are 3.98 mM and 4.94 mM, respectively. The developed thread-based microfluidic system provides a simple yet high performance for clinical diagnostics.

4.
Breast Cancer Res ; 16(3): R57, 2014 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-24890385

RESUMO

INTRODUCTION: Transforming growth factor-ßs (TGF-ßs) play a dual role in breast cancer, with context-dependent tumor-suppressive or pro-oncogenic effects. TGF-ß antagonists are showing promise in early-phase clinical oncology trials to neutralize the pro-oncogenic effects. However, there is currently no way to determine whether the tumor-suppressive effects of TGF-ß are still active in human breast tumors at the time of surgery and treatment, a situation that could lead to adverse therapeutic responses. METHODS: Using a breast cancer progression model that exemplifies the dual role of TGF-ß, promoter-wide chromatin immunoprecipitation and transcriptomic approaches were applied to identify a core set of TGF-ß-regulated genes that specifically reflect only the tumor-suppressor arm of the pathway. The clinical significance of this signature and the underlying biology were investigated using bioinformatic analyses in clinical breast cancer datasets, and knockdown validation approaches in tumor xenografts. RESULTS: TGF-ß-driven tumor suppression was highly dependent on Smad3, and Smad3 target genes that were specifically enriched for involvement in tumor suppression were identified. Patterns of Smad3 binding reflected the preexisting active chromatin landscape, and target genes were frequently regulated in opposite directions in vitro and in vivo, highlighting the strong contextuality of TGF-ß action. An in vivo-weighted TGF-ß/Smad3 tumor-suppressor signature was associated with good outcome in estrogen receptor-positive breast cancer cohorts. TGF-ß/Smad3 effects on cell proliferation, differentiation and ephrin signaling contributed to the observed tumor suppression. CONCLUSIONS: Tumor-suppressive effects of TGF-ß persist in some breast cancer patients at the time of surgery and affect clinical outcome. Carefully tailored in vitro/in vivo genomic approaches can identify such patients for exclusion from treatment with TGF-ß antagonists.


Assuntos
Neoplasias da Mama/genética , Proteína Smad3/genética , Fator de Crescimento Transformador beta/genética , Proteínas Supressoras de Tumor/genética , Neoplasias da Mama/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Efrinas/metabolismo , Feminino , Humanos , Regiões Promotoras Genéticas/genética , Interferência de RNA , RNA Interferente Pequeno , Receptor EphA2/metabolismo , Proteína Smad2/genética , Proteína Smad3/biossíntese , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/biossíntese , Proteínas Supressoras de Tumor/antagonistas & inibidores
5.
PLoS One ; 8(10): e76115, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24146823

RESUMO

The role of myeloid derived suppressor cells (MDSCs) in promoting tumorigenesis is well-established, and significant effort is being made to further characterize surface markers on MDSCs both for better diagnosis and as potential targets for therapy. Here we show that the B cell receptor adaptor molecule CD79a is unexpectedly expressed on immature bone marrow myeloid cells, and is upregulated on MDSCs generated in multiple different mouse models of metastatic but not non-metastatic cancer. CD79a on MDSCs is upregulated and activated in response to soluble factors secreted by tumor cells. Activation of CD79a on mouse MDSCs, by crosslinking with a specific antibody, maintained their immature phenotype (CD11b+Gr1+), enhanced their migration, increased their suppressive effect on T cell proliferation, and increased secretion of pro-tumorigenic cytokines such as IL-6 and CCL22. Furthermore, crosslinking CD79a on myeloid cells activated signaling through Syk, BLNK, ERK and STAT3 phosphorylation. In vivo, CD79+ myeloid cells showed enhanced ability to promote primary tumor growth and metastasis. Finally we demonstrate that CD79a is upregulated on circulating myeloid cells from lung cancer patients, and that CD79a+ myeloid cells infiltrate human breast tumors. We propose that CD79a plays a functional role in the tumor promoting effects of myeloid cells, and may represent a novel target for cancer therapy.


Assuntos
Linfócitos B/metabolismo , Neoplasias da Mama/genética , Antígenos CD79/genética , Carcinogênese/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Anticorpos/metabolismo , Complexo Antígeno-Anticorpo/metabolismo , Linfócitos B/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Antígenos CD79/metabolismo , Carcinogênese/metabolismo , Carcinogênese/patologia , Movimento Celular , Proliferação de Células , Quimiocina CCL22/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Interleucina-6/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Células Mieloides/metabolismo , Células Mieloides/patologia , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Quinase Syk , Linfócitos T/metabolismo , Linfócitos T/patologia
6.
Mol Cancer Res ; 10(10): 1389-99, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22878587

RESUMO

TGF-ß plays a dual role in epithelial carcinogenesis with the potential to either suppress or promote tumor progression. We found that levels of Smad3 mRNA, a critical mediator of TGF-ß signaling, are reduced by approximately 60% in human breast cancer. We therefore used conditionally immortalized mammary epithelial cells (IMEC) of differing Smad3 genotypes to quantitatively address the Smad3 requirement for different biologic responses to TGF-ß. We found that a two-fold reduction in Smad3 gene dosage led to complex effects on TGF-ß responses; the growth-inhibitory response was retained, the pro-apoptotic response was lost, the migratory response was reduced, and the invasion response was enhanced. Loss of the pro-apoptotic response in the Smad3(+/-) IMECs correlated with loss of Smad3 binding to the Bcl-2 locus, whereas retention of the growth-inhibitory response in Smad3 IMECs correlated with retention of Smad3 binding to the c-Myc locus. Addressing the integrated outcome of these changes in vivo, we showed that reduced Smad3 levels enhanced metastasis in two independent models of metastatic breast cancer. Our results suggest that different biologic responses to TGF-ß in the mammary epithelium are differentially affected by Smad3 dosage and that a mere two-fold reduction in Smad3 is sufficient to promote metastasis.


Assuntos
Neoplasias da Mama/patologia , Epitélio/metabolismo , Dosagem de Genes/efeitos dos fármacos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Humanas/metabolismo , Proteína Smad3/genética , Fator de Crescimento Transformador beta/farmacologia , Animais , Neoplasias da Mama/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Progressão da Doença , Elementos Facilitadores Genéticos/genética , Epitélio/efeitos dos fármacos , Epitélio/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Humanas/efeitos dos fármacos , Glândulas Mamárias Humanas/patologia , Camundongos , Metástase Neoplásica , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína Smad3/metabolismo
7.
Breast Cancer Res ; 12(5): R83, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20942910

RESUMO

INTRODUCTION: Molecular dissection of the signaling pathways that underlie complex biological responses in the mammary epithelium is limited by the difficulty of propagating large numbers of mouse mammary epithelial cells, and by the inability of ribonucleic acid interference (RNAi)-based knockdown approaches to fully ablate gene function. Here we describe a method for the generation of conditionally immortalized mammary epithelial cells with defined genetic defects, and we show how such cells can be used to investigate complex signal transduction processes using the transforming growth factor beta (TGFß/Smad pathway as an example. METHODS: We intercrossed the previously described H-2Kb-tsA58 transgenic mouse (Immortomouse) which expresses a temperature-sensitive mutant of the simian virus-40 large T-antigen (tsTAg), with mice of differing Smad genotypes. A panel of conditionally immortalized mammary epithelial cell (IMEC) cultures were derived from the virgin mammary glands of offspring of these crosses and used to assess the Smad dependency of different biological responses to TGFß. RESULTS: IMECs could be propagated indefinitely at permissive temperatures and had a stable epithelial phenotype, resembling primary mammary epithelial cells with respect to several criteria, including responsiveness to TGFß. Using this panel of cells, we demonstrated that Smad3, but not Smad2, is necessary for TGFß-induced apoptotic, growth inhibitory and EMT responses, whereas either Smad can support TGFß-induced invasion as long as a threshold level of total Smad is exceeded. CONCLUSIONS: This work demonstrates the practicality and utility of generating conditionally immortalized mammary epithelial cell lines from genetically modified Immortomice for detailed investigation of complex signaling pathways in the mammary epithelium.


Assuntos
Células Epiteliais/metabolismo , Glândulas Mamárias Animais/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Apoptose/genética , Diferenciação Celular , Movimento Celular , Células Cultivadas , Transição Epitelial-Mesenquimal/genética , Feminino , Técnicas de Inativação de Genes , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Transgênicos , Proteína Smad2/genética , Proteína Smad3/genética , Fator de Crescimento Transformador beta/genética
8.
Cancer Lett ; 287(2): 172-81, 2010 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-19586713

RESUMO

The cancer stem cell (CSC) hypothesis proposes that CSCs are the root of cancer and cause cancer metastasis and recurrence. In this study, we examined whether Ras signaling is associated with stemness of the CSCs population characterized by the stem cell antigen (Sca-1) phenotype in a 4T1 syngeneic mouse model of breast cancer. The Sca-1(pos) putative CSCs had high levels of activated Ras and phosphorylated MEK (p-MEK), compared with counterparts. The Ras farnesylation inhibitor (FTI-277) suppressed the maintenance and expansion of CSCs. Therefore, selective inhibition of Ras activation may be useful for stem-specific cancer therapy.


Assuntos
Antígenos Ly/metabolismo , Neoplasias da Mama/metabolismo , Proliferação de Células , Proteínas de Membrana/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas ras/metabolismo , Aldeído Desidrogenase/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Farnesiltranstransferase/antagonistas & inibidores , Farnesiltranstransferase/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , MAP Quinase Quinase Quinases/metabolismo , Metionina/análogos & derivados , Metionina/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Fosforilação , Prenilação de Proteína , Transdução de Sinais , Esferoides Celulares
9.
Cancer Res ; 69(3): 735-40, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19155312

RESUMO

Controlled protein degradation mediated by ubiquitin/proteasome system (UPS) plays a crucial role in modulating a broad range of cellular responses. Dysregulation of the UPS often accompanies tumorigenesis and progression. Here, we report that Smad ubiquitination regulatory factor 2 (Smurf2), a HECT-domain containing E3 ubiquitin ligase, is up-regulated in certain breast cancer tissues and cells. We show that reduction of Smurf2 expression with specific short interfering RNA in metastatic breast cancer cells induces cell rounding and reorganization of the actin cytoskeleton, which are associated with a less motile and invasive phenotype. Overexpression of Smurf2 promotes metastasis in a nude mouse model and increases migration and invasion of breast cancer cells. Moreover, expression of Smurf2CG, an E3 ligase-defective mutant of Smurf2, suppresses the above metastatic behaviors. These results establish an important role for Smurf2 in breast cancer progression and indicate that Smurf2 is a novel regulator of breast cancer cell migration and invasion.


Assuntos
Neoplasias da Mama/enzimologia , Movimento Celular/fisiologia , Ubiquitina-Proteína Ligases/biossíntese , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia
10.
Cancer Res ; 68(10): 3915-23, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18483277

RESUMO

Overexpression of the immunosuppressive cytokine transforming growth factor beta (TGF-beta) is one strategy that tumors have developed to evade effective immunesurveillance. Using transplantable models of breast and colon cancer, we made the unexpected finding that CD8+ cells in tumor-bearing animals can directly promote tumorigenesis, by a mechanism that is dependent on TGF-beta. We showed that CD8+ splenocytes from tumor-bearing mice expressed elevated interleukin (IL)-17 when compared with naive mice, and that CD8+ T cells could be induced to make IL-17 on addition of TGF-beta and IL-6 in vitro. Treatment of mice with anti-TGF-beta antibodies in vivo reduced IL-17 expression both in the tumor and the locoregional lymph nodes. Although IL-17 has not previously been shown to act as a survival factor for epithelial cells, we found that IL-17 suppressed apoptosis of several tumor cell lines in vitro, suggesting that this altered T-cell polarization has the potential to promote tumorigenesis directly, rather than indirectly through inflammatory sequelae. Consistent with this hypothesis, knockdown of the IL-17 receptor in 4T1 mouse mammary cancer cells enhanced apoptosis and decreased tumor growth in vivo. Thus, in addition to suppressing immune surveillance, tumor-induced TGF-beta may actively subvert the CD8+ arm of the immune system into directly promoting tumor growth by an IL-17-dependent mechanism.


Assuntos
Regulação Neoplásica da Expressão Gênica , Interleucina-17/fisiologia , Neoplasias/imunologia , Neoplasias/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Ratos
11.
Cancer Cell ; 9(6): 445-57, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16766264

RESUMO

In the liver, derangement of TGF-beta signaling is associated with an increased incidence of hepatocellular carcinoma (HCC), but the mechanism is not clear. We report here that forced expression of a major TGF-beta signaling transducer, Smad3, reduces susceptibility to HCC in a chemically induced murine model. This protection is conferred by Smad3's ability to promote apoptosis by repressing Bcl-2 transcription in vivo through a GC-rich element in the Bcl-2 promoter. We also show that the proapoptotic activity of Smad3 requires both input from TGF-beta signaling and activation of p38 MAPK, which occurs selectively in the liver tumor cells. Thus, Smad3 enables the tumor suppression function of TGF-beta by serving as a physiological mediator of TGF-beta-induced apoptosis.


Assuntos
Apoptose , Carcinoma Hepatocelular/metabolismo , Hepatócitos/patologia , Neoplasias Hepáticas Experimentais/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Proteína Smad3/fisiologia , Animais , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/patologia , Células Cultivadas , Suscetibilidade a Doenças , Regulação para Baixo , Hepatócitos/metabolismo , Neoplasias Hepáticas Experimentais/induzido quimicamente , Neoplasias Hepáticas Experimentais/patologia , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Transporte Proteico , Proteínas Proto-Oncogênicas c-bcl-2/genética , Transdução de Sinais , Proteína Smad3/biossíntese , Proteína Smad3/genética , Transcrição Gênica , Fator de Crescimento Transformador beta/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Exp Cell Res ; 279(1): 80-90, 2002 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-12213216

RESUMO

Activation of fatty acid synthase (FAS) expression and fatty acid synthesis is a common event in tumor cells from a variety of human cancers and is closely linked to malignant transformation and to tumor virulence in population studies of human cancer. We now show that, in contrast to nutritional regulation of lipogenesis in liver or adipose tissue, changes in fatty acid metabolism during in vitro transformation of the human mammary epithelial cell line MCF-10a are driven by increases in epidermal growth factor signaling, acting in major part through the mitogen-activated protein (MAP) kinase and phosphatidylinositol (PI) 3-kinase signaling cascades. H-ras transformation of MCF-10a cells resulted in upregulation of MAP kinase and PI 3-kinase signals, upregulation of sterol regulatory element binding protein 1 (SREBP-1) transcription factor levels, and upregulation of FAS expression and FA synthesis. Deletion of the major SREBP binding site from the FAS promoter abrogated transcription in transformed MCF-10a cells. Inhibitors of MAP and PI 3-kinases downregulated SREBP-1 levels and decreased transcription from the FAS promoter, reducing FAS expression and fatty acid synthesis in transformed MCF-10a cells and in MCF-7 and HCT116 carcinoma cells. H-ras transformation sensitized MCF-10a cells to the FAS inhibitors cerulenin and C-75. These results confirm an important role for SREBP-1 in neoplastic lipogenesis, and provide a likely basis for the linkage of upregulated fatty acid metabolism with neoplastic transformation and with tumor virulence, since MAP and PI 3-kinase signaling contributes to both.


Assuntos
Mama/metabolismo , Carcinoma/metabolismo , Ácidos Graxos/biossíntese , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Fatores de Transcrição , Mama/citologia , Mama/enzimologia , Proteínas Estimuladoras de Ligação a CCAAT/biossíntese , Carcinoma/enzimologia , Linhagem Celular Transformada , Transformação Celular Neoplásica , Proteínas de Ligação a DNA/biossíntese , Inibidores Enzimáticos/farmacologia , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Células Epiteliais/metabolismo , Ácido Graxo Sintases/antagonistas & inibidores , Ácido Graxo Sintases/metabolismo , Inibidores do Crescimento/farmacologia , Humanos , Metabolismo dos Lipídeos , MAP Quinase Quinase 1 , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 1 , Células Tumorais Cultivadas , Proteínas ras/fisiologia
13.
J Clin Invest ; 109(12): 1607-15, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12070308

RESUMO

TGF-betas play diverse and complex roles in many biological processes. In tumorigenesis, they can function either as tumor suppressors or as pro-oncogenic factors, depending on the stage of the disease. We have developed transgenic mice expressing a TGF-beta antagonist of the soluble type II TGF-beta receptor:Fc fusion protein class, under the regulation of the mammary-selective MMTV-LTR promoter/enhancer. Biologically significant levels of antagonist were detectable in the serum and most tissues of this mouse line. The mice were resistant to the development of metastases at multiple organ sites when compared with wild-type controls, both in a tail vein metastasis assay using isogenic melanoma cells and in crosses with the MMTV-neu transgenic mouse model of metastatic breast cancer. Importantly, metastasis from endogenous mammary tumors was suppressed without any enhancement of primary tumorigenesis. Furthermore, aged transgenic mice did not exhibit the severe pathology characteristic of TGF-beta null mice, despite lifetime exposure to the antagonist. The data suggest that in vivo the antagonist may selectively neutralize the undesirable TGF-beta associated with metastasis, while sparing the regulatory roles of TGF-betas in normal tissues. Thus this soluble TGF-beta antagonist has potential for long-term clinical use in the prevention of metastasis.


Assuntos
Fragmentos Fc das Imunoglobulinas/fisiologia , Imunoglobulina G/fisiologia , Neoplasias Hepáticas/secundário , Neoplasias Mamárias Animais/prevenção & controle , Melanoma Experimental/prevenção & controle , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Feminino , Vetores Genéticos , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Imunoglobulina G/genética , Neoplasias Mamárias Animais/patologia , Vírus do Tumor Mamário do Camundongo , Camundongos , Camundongos Transgênicos , Metástase Neoplásica/prevenção & controle , Proteínas Serina-Treonina Quinases , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/fisiologia , Solubilidade , Células Tumorais Cultivadas
14.
Cell Growth Differ ; 13(3): 123-30, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11959813

RESUMO

Transforming growth factor-beta (TGF-beta) regulates proliferation, morphogenesis, and functional differentiation in the mammary gland and plays complex roles in mammary tumorigenesis. Here we show that the signaling mediators Smad1-Smad5 are expressed at all stages of mammary gland development. To begin to investigate which Smads mediate which TGF-beta responses, we have analyzed mammary gland development in Smad3 null mice. Smad3 null virgin females showed delayed mammary gland development. However, this phenotype was secondary to ovarian insufficiency because Smad3 null mammary epithelium developed normally in hormonally supplemented Smad3 null mice or when transplanted into wild-type hosts. Absence of Smad3 had no effect on the ability of TGF-beta to inhibit the growth of mammary epithelial cells in culture, and no compensatory changes in expression or activation of Smad2 were seen in the Smad3 null epithelium. A small but significant decrease in apoptotic cells was seen in involuting glands from Smad3 null transplants. The results suggest that epithelial Smad3 is dispensable for TGF-beta effects on proliferation and differentiation in the mammary gland, but that it contributes in a nonredundant manner to the induction of apoptosis.


Assuntos
Apoptose , Proteínas de Ligação a DNA/fisiologia , Glândulas Mamárias Animais/fisiologia , Transativadores/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Animais , Diferenciação Celular , Divisão Celular , Proteínas de Ligação a DNA/metabolismo , Epitélio/fisiologia , Feminino , Hormônios/fisiologia , Lactação , Masculino , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Ovário/citologia , Ovário/fisiologia , Proteínas Smad , Proteína Smad1 , Proteína Smad2 , Proteína Smad3 , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA