Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Mol Cell Biol ; 19(12): 8169-79, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10567542

RESUMO

The Grb2 and Shc adapter proteins play critical roles in coupling activated growth factor receptors to several cellular signaling pathways. To assess the role of these molecules in mammary epithelial development and tumorigenesis, we have generated transgenic mice which individually express the Grb2 and Shc proteins in the mammary epithelium. Although mammary epithelial cell-specific expression of Grb2 or Shc accelerated ductal morphogenesis, mammary tumors were rarely observed in these strains. To explore the potential role of these adapter proteins in mammary tumorigenesis, mice coexpressing either Shc or Grb2 and a mutant form of polyomavirus middle T (PyV mT) antigen in the mammary epithelium were generated. Coexpression of either Shc or Grb2 with the mutant PyV mT antigen resulted in a dramatic acceleration of mammary tumorigenesis compared to parental mutant PyV mT strain. The increased rate of tumor formation observed in these mice was correlated with activation of the epidermal growth factor receptor family and mitogen-activated protein kinase pathway. These observations suggest that elevated levels of the Grb2 or Shc adapter protein can accelerate mammary tumor progression by sensitizing the mammary epithelial cell to growth factor receptor signaling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Antígenos Transformantes de Poliomavirus/fisiologia , Neoplasias Mamárias Experimentais/patologia , Proteínas/fisiologia , Animais , Antígenos Transformantes de Poliomavirus/genética , Feminino , Proteína Adaptadora GRB2 , Expressão Gênica , Vetores Genéticos , Humanos , Vírus do Tumor Mamário do Camundongo , Camundongos , Camundongos Transgênicos , Morfogênese , Biossíntese de Proteínas , Proteínas/genética , Coelhos , Proteínas Adaptadoras da Sinalização Shc , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
2.
Oncogene ; 36(25): 3553-3561, 2017 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-28135251

RESUMO

Current therapeutic outcomes for breast cancer underscore the complexity of treating a heterogeneous disease. Indeed, studies have shown that differences in gene expression among patients with the same subtype of breast cancer are correlated with the response to treatment. This strongly suggests that there is an urgent need to treat breast cancer with a personalized approach. Here we employed cell signaling pathway signatures to predict pathway activity in subtypes of MMTV-Myc mammary tumors. We then split tumors into subsets and developed individualized combinatorial treatments for two subtypes with distinct pathway activation patterns. Elevation of the EGFR, RAS and TGFß pathways was observed in one subtype whereas these pathways were not predicted to be active in the other subtype that had high predicted activity of the Myc, Stat3 and Akt pathways. In a proof-of-principle experiment, treatment of these two subtypes with targeted therapies inhibited tumor growth only in the subtype of tumor where the therapy was designed to be active. We then analyzed gene expression profiles of human breast cancer patients and patient-derived xenograft (PDX) samples to predict pathway activity, and validated our approach of developing individualized treatments in mice with PDX tumors. Importantly, our combinatorial therapy resulted in tumor regression, including regression in PDX samples from triple-negative breast cancer. Together our data is a proof-of-principle experiment that demonstrates that cell signaling pathway signature-guided treatment for breast cancer is viable.


Assuntos
Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Medicina de Precisão , Transdução de Sinais , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Feminino , Perfilação da Expressão Gênica , Xenoenxertos , Humanos , Camundongos , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos
3.
Oncogene ; 35(47): 6053-6064, 2016 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-27157621

RESUMO

Amplification and overexpression of erbB2/neu proto-oncogene is observed in 20-30% human breast cancer and is inversely correlated with the survival of the patient. Despite this, somatic activating mutations within erbB2 in human breast cancers are rare. However, we have previously reported that a splice isoform of erbB2, containing an in-frame deletion of exon 16 (herein referred to as ErbB2ΔEx16), results in oncogenic activation of erbB2 because of constitutive dimerization of the ErbB2 receptor. Here, we demonstrate that the ErbB2ΔEx16 is a major oncogenic driver in breast cancer that constitutively signals from the cell surface. We further show that inducible expression of the ErbB2ΔEx16 variant in mammary gland of transgenic mice results in the rapid development of metastatic multifocal mammary tumors. Genetic and biochemical characterization of the ErbB2ΔEx16-derived mammary tumors exhibit several unique features that distinguish this model from the conventional ErbB2 ones expressing the erbB2 proto-oncogene in mammary epithelium. Unlike the wild-type ErbB2-derived tumors that express luminal keratins, ErbB2ΔEx16-derived tumors exhibit high degree of intratumoral heterogeneity co-expressing both basal and luminal keratins. Consistent with these distinct pathological features, the ErbB2ΔEx16 tumors exhibit distinct signaling and gene expression profiles that correlate with activation of number of key transcription factors implicated in breast cancer metastasis and cancer stem cell renewal.


Assuntos
Processamento Alternativo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Transformação Celular Neoplásica/genética , Receptor ErbB-2/genética , Microambiente Tumoral/genética , Animais , Linhagem Celular Tumoral , Análise por Conglomerados , Modelos Animais de Doenças , Éxons , Matriz Extracelular/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Transgênicos , Metástase Neoplásica , Fenótipo , Proto-Oncogene Mas , Deleção de Sequência , Fatores de Transcrição/metabolismo
4.
Oncogene ; 34(2): 217-25, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-24362522

RESUMO

HER2/Neu is amplified and overexpressed in a large proportion of human breast cancers, but the signaling pathways that contribute to tumor development and metastatic progression are not completely understood. Using gene expression data and pathway signatures, we predicted a role for activator E2F transcription factors in Neu-induced tumors. This was genetically tested by interbreeding Neu transgenics with knockouts of the three activator E2Fs. Loss of any E2F delayed Neu-induced tumor onset. E2F1 loss accelerated tumor growth, while E2F2 and E2F3 loss did not. Strikingly, it was observed that loss of E2F1 or E2F2 significantly reduced the metastatic capacity of the tumor and this was associated with a reduction in circulating tumor cells in the E2F2 knockout. Gene expression analysis between the tumors in the various E2F-mutant backgrounds revealed that there was extensive compensation by other E2F family members in the individual knockouts, underscoring the importance of the E2Fs in HER2/Neu-induced tumors. Extension to HER2-positive (HER2+) human breast cancer revealed a number of HER2+ subtypes based on E2F activity with differences in relapse-free survival times. Taken together, these data demonstrate that the E2F transcription factors are integral to HER2+ tumor development and progression.


Assuntos
Neoplasias da Mama/metabolismo , Fatores de Transcrição E2F/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Receptor ErbB-2/metabolismo , Animais , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Carcinogênese , Modelos Animais de Doenças , Fatores de Transcrição E2F/deficiência , Fatores de Transcrição E2F/genética , Feminino , Humanos , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Knockout , Camundongos Nus , Metástase Neoplásica , Células Neoplásicas Circulantes/patologia , Transdução de Sinais
5.
Oncogene ; 32(10): 1296-304, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-22525269

RESUMO

Expression of c-Myc is highly prevalent in human breast cancer and stability of the oncoprotein is regulated through Ras-regulated phosphorylation at serine 62 and threonine 58. Previous studies have illustrated the importance of accumulation of KRas mutations in Myc-mediated tumor formation. To examine Myc dependence upon Ras mutations we have generated MMTV regulated Myc and Myc T58A transgenic mice. Expression of the more stable T58A Myc allele resulted in a reduction in KRas-activating mutations. However, in a low-level expression T58A Myc transgenic, the majority of the tumors were squamous or epithelial-to-mesenchymal in nature and accumulated KRas mutations at a higher frequency. Interestingly, we show that these mice develop similar gene expression patterns and signaling pathway utilization as a subtype of human claudin-low breast cancer. Indeed, our results demonstrate a clear division in human claudin-low tumors based on Myc pathway activation and target genes. Together, our results demonstrate that Myc expression and stability has critical effects on molecular heterogeneity in mouse mammary tumors that parallel subtypes of human breast cancer.


Assuntos
Neoplasias da Mama/genética , Claudinas/metabolismo , Genes myc , Genes ras , Neoplasias Mamárias Experimentais/genética , Mutação , Animais , Neoplasias da Mama/patologia , Diferenciação Celular/genética , Processos de Crescimento Celular/genética , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Transdução de Sinais
6.
Oncogene ; 31(20): 2545-54, 2012 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21996730

RESUMO

A hallmark of human cancer is heterogeneity, reflecting the complex series of changes resulting in the activation of oncogenes coupled with inactivation of tumor suppressor genes. Breast cancer is no exception and indeed, many studies have revealed considerable complexity and heterogeneity in the population of primary breast tumors and substantial changes in a recurrent breast tumor that has acquired metastatic properties and drug resistance. We have made use of a Myc-inducible transgenic mouse model of breast cancer in which elimination of Myc activity following tumor development initially leads to a regression of a subset of tumors generally followed by de novo Myc-independent growth. We have observed that tumors that grow independent of Myc expression have gene profiles that are distinct from the primary tumors with characteristics indicative of an epithelial-mesenchymal transition (EMT) phenotype. Phenotypic analyses of Myc-independent tumors confirm the acquisition of an EMT phenotype suggested to be associated with invasive and migratory properties in human cancer cells. Further genomic analyses reveal mouse mammary tumors growing independent of myc have a higher probability of exhibiting a gene signature similar to that observed for human 'tumor-initiating' cells. Collectively, the data reveal genetic alterations that underlie tumor progression and an escape from Myc-dependent growth in a transgenic mouse model that can provide insights to what occurs in human cancers as they acquire drug resistance and metastatic properties.


Assuntos
Neoplasias da Mama , Transformação Celular Neoplásica , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal/genética , Neoplasias Mamárias Experimentais , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes ras , Humanos , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Camundongos , Células-Tronco Neoplásicas/patologia , Fator de Crescimento Transformador alfa/genética , Fator de Crescimento Transformador beta/genética
7.
Oncogene ; 28(31): 2796-805, 2009 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-19483725

RESUMO

The oncogenic phenotype is complex, resulting from the accumulation of multiple somatic mutations that lead to the deregulation of growth regulatory and cell fate controlling activities and pathways. The ability to dissect this complexity, so as to reveal discrete aspects of the biology underlying the oncogenic phenotype, is critical to understanding the various mechanisms of disease as well as to reveal opportunities for novel therapeutic strategies. Previous work has characterized the process of anchorage-independent growth of cancer cells in vitro as a key aspect of the tumor phenotype, particularly with respect to metastatic potential. Nevertheless, it remains a major challenge to translate these cell biology findings into the context of human tumors. We previously used DNA microarray assays to develop expression signatures, which have the capacity to identify subtle distinctions in biological states and can be used to connect in vitro and in vivo states. Here we describe the development of a signature of anchorage-independent growth, show that the signature exhibits characteristics of deregulated mitochondrial function and then demonstrate that the signature identifies human tumors with the potential for metastasis.


Assuntos
Proliferação de Células , Perfilação da Expressão Gênica , Neoplasias/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Células 3T3 , Animais , Teorema de Bayes , Adesão Celular , Linhagem Celular Tumoral , Metabolismo Energético , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Metástase Neoplásica , Neoplasias/metabolismo , Neoplasias/patologia , Fenótipo , Prognóstico , Análise de Regressão , Ensaio Tumoral de Célula-Tronco
8.
Breast Cancer Res ; 2(3): 211-6, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11250712

RESUMO

The ability of growth factors and their cognate receptors to induce mammary epithelial proliferation and differentiation is dependent on their ability to activate a number of specific signal transduction pathways. Aberrant expression of specific receptor tyrosine kinases (RTKs) has been implicated in the genesis of a significant proportion of sporadic human breast cancers. Indeed, mammary epithelial expression of activated RTKs such as ErbB2/neu in transgenic mice has resulted in the efficient induction of metastatic mammary tumours. Although it is clear from these studies that activation these growth factor receptor signalling cascades are directly involved in mammary tumour progression, the precise interaction of each of these signalling pathways in mammary tumourigenesis and metastasis remains to be elucidated. The present review focuses on the role of several specific signalling pathways that have been implicated as important components in RTK-mediated signal transduction. In particular, it focuses on two well characterized transgenic breast cancer models that carry the polyomavirus middle T(PyV mT) and neu oncogenes.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias Mamárias Experimentais/genética , Camundongos , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Animais , Antígenos Transformantes de Poliomavirus/genética , Neoplasias da Mama/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Mamárias Experimentais/metabolismo , Camundongos Transgênicos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Tirosina Quinases/genética , Proto-Oncogenes , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Transdução de Sinais/genética , Ativação Transcricional , Proteínas ras/metabolismo , Quinases da Família src/metabolismo
9.
Proc Natl Acad Sci U S A ; 97(7): 3444-9, 2000 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-10716706

RESUMO

The neu (c-erbB-2, Her-2) protooncogene is amplified and overexpressed in 20-30% of human breast cancers. Although transgenic mouse models have illustrated the role of Neu in the induction of mammary tumors, Neu expression in these models is driven by a strong viral promoter of questionable relevance to the human disease. To ascertain whether expression of activated Neu under the control of the endogenous promoter in the mammary gland could induce mammary tumors we have generated mice that conditionally express activated Neu under the transcriptional control of the intact endogenous Neu promoter. Expression of oncogenic neu in the mammary gland resulted in accelerated lobulo-alveolar development and formation of focal mammary tumors after a long latency period. However, expression of activated Neu under the normal transcriptional control of the endogenous promoter was not sufficient for the initiation of mammary carcinogenesis. Strikingly, all mammary tumors bear amplified copies (2-22 copies) of the activated neu allele relative to the wild-type allele and express highly elevated levels of neu transcript and protein. Thus, like human erbB-2-positive breast tumors, mammary tumorigenesis in this mouse model requires the amplification and commensurate elevated expression of the neu gene.


Assuntos
Amplificação de Genes , Genes erbB-2 , Neoplasias Mamárias Experimentais/genética , Animais , DNA Complementar , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA