Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Res ; 80(19): 4071-4086, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32631905

RESUMO

The complex yet interrelated connections between cancer metabolism, gene expression, and oncogenic driver genes have the potential to identify novel biomarkers and drug targets with prognostic and therapeutic value. Here we effectively integrated metabolomics and gene expression data from breast cancer mouse models through a novel unbiased correlation-based network analysis. This approach identified 35 metabolite and 34 gene hubs with the most network correlations. These hubs have prognostic value and are likely integral to tumor metabolism and breast cancer. The gene hub Aquaporin-7 (Aqp7), a water and glycerol channel, was identified as a novel regulator of breast cancer. AQP7 was prognostic of overall survival in patients with breast cancer. In mouse breast cancer models, reduced expression of Aqp7 caused reduced primary tumor burden and lung metastasis. Metabolomics and complex lipid profiling of cells and tumors with reduced Aqp7 revealed significantly altered lipid metabolism, glutathione metabolism, and urea/arginine metabolism compared with controls. These data identify AQP7 as a critical regulator of metabolic and signaling responses to environmental cellular stresses in breast cancer, highlighting AQP7 as a potential cancer-specific therapeutic vulnerability. SIGNIFICANCE: Aquaporin-7 is identified as a critical regulator of nutrient availability and signaling that responds to cellular stresses, making it an attractive therapeutic target in breast cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/19/4071/F1.large.jpg.


Assuntos
Aquaporinas/genética , Aquaporinas/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Adipócitos/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Metabolismo dos Carboidratos , Movimento Celular , Feminino , Regulação Neoplásica da Expressão Gênica , Glicolipídeos/metabolismo , Glicólise , Humanos , Inositol/análogos & derivados , Inositol/metabolismo , Lipídeos/biossíntese , Lipídeos/genética , Neoplasias Pulmonares/secundário , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/fisiologia , Óxido Nítrico/metabolismo , Prognóstico
2.
Cancer Res ; 80(5): 1156-1170, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31932454

RESUMO

The noncanonical Wnt ligand Wnt5a is found in high concentrations in ascites of women with ovarian cancer. In this study, we elucidated the role of Wnt5a in ovarian cancer metastasis. Wnt5a promoted ovarian tumor cell adhesion to peritoneal mesothelial cells as well as migration and invasion, leading to colonization of peritoneal explants. Host components of the ovarian tumor microenvironment, notably peritoneal mesothelial cells and visceral adipose, secreted Wnt5a. Conditional knockout of host WNT5A significantly reduced peritoneal metastatic tumor burden. Tumors formed in WNT5A knockout mice had elevated cytotoxic T cells, increased M1 macrophages, and decreased M2 macrophages, indicating that host Wnt5a promotes an immunosuppressive microenvironment. The Src family kinase Fgr was identified as a downstream effector of Wnt5a. These results highlight a previously unreported role for host-expressed Wnt5a in ovarian cancer metastasis and suggest Fgr as a novel target for inhibition of ovarian cancer metastatic progression.Significance: This study establishes host-derived Wnt5a, expressed by peritoneal mesothelial cells and adipocytes, as a primary regulator of ovarian cancer intraperitoneal metastatic dissemination and identifies Fgr kinase as novel target for inhibition of metastasis.


Assuntos
Carcinoma Epitelial do Ovário/patologia , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/imunologia , Peritônio/patologia , Proteínas Proto-Oncogênicas/metabolismo , Proteína Wnt-5a/metabolismo , Quinases da Família src/metabolismo , Animais , Carcinoma Epitelial do Ovário/imunologia , Adesão Celular/imunologia , Linhagem Celular Tumoral , Movimento Celular/imunologia , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/patologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Macrófagos/imunologia , Camundongos , Camundongos Knockout , Metástase Neoplásica/imunologia , Metástase Neoplásica/patologia , Neoplasias Ovarianas/imunologia , Neoplasias Peritoneais/secundário , Peritônio/citologia , Peritônio/imunologia , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/imunologia , Microambiente Tumoral/imunologia , Proteína Wnt-5a/genética , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Mol Cancer Res ; 12(6): 855-66, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24803643

RESUMO

UNLABELLED: Carcinoma-associated fibroblasts (CAFs) are now widely appreciated for their contributions to tumor progression. However, the ability of CAFs to regulate anoikis, detachment-induced cell death, has yet to be investigated. Here, a new role for CAFs in blocking anoikis in multiple cell lines, facilitating luminal filling in three-dimensional cell culture, and promoting anchorage-independent growth is defined. In addition, a novel mechanism underlying anoikis inhibition is discovered. Importantly, it was demonstrated that CAFs secrete elevated quantities of insulin-like growth factor-binding proteins (IGFBPs) that are both necessary for CAF-mediated anoikis inhibition and sufficient to block anoikis in the absence of CAFs. Furthermore, these data reveal a unique antiapoptotic mechanism for IGFBPs: the stabilization of the antiapoptotic protein Mcl-1. In aggregate, these data delineate a novel role for CAFs in promoting cell survival during detachment and unveil an additional mechanism by which the tumor microenvironment contributes to cancer progression. These results also identify IGFBPs as potential targets for the development of novel chemotherapeutics designed to eliminate detached cancer cells. IMPLICATIONS: The ability of CAF-secreted IGFBPs to block anoikis in breast cancer represents a novel target for the development of therapeutics aimed at specifically eliminating extracellular matrix-detached breast cancer cells.


Assuntos
Anoikis/fisiologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Fibroblastos/metabolismo , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Feminino , Xenoenxertos , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Camundongos , Camundongos Nus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA