Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Int J Cancer ; 131(7): 1537-48, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22262494

RESUMO

Membrane-type 4 matrix metalloprotease (MT4-MMP) expression in breast adenocarcinoma stimulates tumor growth and metastatic spreading to the lung. However, whether these pro-tumorigenic and pro-metastatic effects of MT4-MMP are related to a proteolytic action is not yet known. Through site directed mutagenesis MT4-MMP has been inactivated in cancer cells through Glutamic acid 249 substitution by Alanine in the active site. Active MT4-MMP triggered an angiogenic switch at day 7 after tumor implantation and drastically accelerated subcutaneous tumor growth as well as lung colonization in recombination activating gene-1-deficient mice. All these effects were abrogated upon MT4-MMP inactivation. In sharp contrast to most MMPs being primarily of stromal origin, we provide evidence that tumor-derived MT4-MMP, but not host-derived MT4-MMP contributes to angiogenesis. A genetic approach using MT4-MMP-deficient mice revealed that the status of MT4-MMP produced by host cells did not affect the angiogenic response. Despite of this tumor intrinsic feature, to exert its tumor promoting effect, MT4-MMP requires a permissive microenvironment. Indeed, tumor-derived MT4-MMP failed to circumvent the lack of an host angio-promoting factor such as plasminogen activator inhibitor-1. Overall, our study demonstrates the key contribution of MT4-MMP catalytic activity in the tumor compartment, at the interface with host cells. It identifies MT4-MMP as a key intrinsic tumor cell determinant that contributes to the elaboration of a permissive microenvironment for metastatic dissemination.


Assuntos
Metaloproteinase 17 da Matriz/metabolismo , Neoplasias/enzimologia , Neoplasias/patologia , Neovascularização Patológica/enzimologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Progressão da Doença , Ativação Enzimática/genética , Humanos , Metaloproteinase 17 da Matriz/genética , Metástase Neoplásica , Neoplasias/genética , Neovascularização Patológica/genética , Proteólise , Microambiente Tumoral/genética
2.
Sci Rep ; 12(1): 7304, 2022 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-35508525

RESUMO

Viral myocarditis (VM) is an important cause of heart failure (HF) in children and adults. However, the molecular determinants involved in cardiac inflammation and cardiomyocyte necrosis remain poorly characterized, and cardioprotective molecules are currently missing. Here, we applied an in vivo method based on the functional selection (FunSel) of cardioprotective factors using AAV vectors for the unbiased identification of novel immunomodulatory molecules in a Coxsackievirus B3 (CVB3)-induced myocarditis mouse model. Two consecutive rounds of in vivo FunSel using an expression library of 60 cytokines were sufficient to identify five cardioprotective factors (IL9, IL3, IL4, IL13, IL15). The screening also revealed three cytokines (IL18, IL17b, and CCL11) that were counter-selected and likely to exert a detrimental effect. The pooled overexpression of the five most enriched cytokines using AAV9 vectors decreased inflammation and reduced cardiac dilatation, persisting at 1 month after treatment. Individual overexpression of IL9, the top ranking in our functional selection, markedly reduced cardiac inflammation and injury, concomitant with an increase of anti-inflammatory Th2-cells and a reduction of pro-inflammatory Th17- and Th22-cells at 14 days post-infection. AAV9-mediated FunSel cardiac screening identified IL9 and other four cytokines (IL3, IL4, IL13, and IL15) as cardioprotective factors in CVB3-induced VM in mice.


Assuntos
Infecções por Coxsackievirus , Miocardite , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Enterovirus Humano B , Inflamação , Interleucina-13 , Interleucina-15 , Interleucina-4 , Interleucina-9 , Camundongos , Camundongos Endogâmicos BALB C , Miocardite/genética
3.
J Cell Mol Med ; 13(9B): 4002-13, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19426156

RESUMO

The present study aims at investigating the mechanism by which membrane-type 4 matrix metalloproteinase (MT4-MMP), a membrane-anchored MMP expressed by human breast tumour cells promotes the metastatic dissemination into lung. We applied experimental (intravenous) and spontaneous (subcutaneous) models of lung metastasis using human breast adenocarcinoma MDA-MB-231 cells overexpressing or not MT4-MMP. We found that MT4-MMP does not affect lymph node colonization nor extravasation of cells from the bloodstream, but increases the intravasation step leading to metastasis. Ultrastructural and fluorescent microscopic observations coupled with automatic computer-assisted quantifications revealed that MT4-MMP expression induces blood vessel enlargement and promotes the detachment of mural cells from the vascular tree, thus causing an increased tumour vascular leak. On this basis, we propose that MT4-MMP promotes lung metastasis by disturbing the tumour vessel integrity and thereby facilitating tumour cell intravasation.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias Pulmonares/enzimologia , Metaloproteinase 17 da Matriz/metabolismo , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/secundário , Metástase Linfática , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/enzimologia , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Nus , Microscopia Eletrônica de Transmissão/métodos , Microscopia de Fluorescência/métodos , Transplante de Neoplasias , Neovascularização Patológica , Pericitos/metabolismo
5.
FEBS J ; 283(4): 704-22, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26663028

RESUMO

Membrane type 4 matrix metalloproteinase (MT4-MMP) [matrix metalloproteinase (MMP) 17] is a GPI-anchored membrane-type MMP expressed on the cell surface of human breast cancer cells. In triple-negative breast cancer cells, MT4-MMP promotes primary tumour growth and lung metastases. Although the trafficking and internalization of the transmembrane membrane type 1 MMP have been extensively investigated, little is known about the regulatory mechanisms of the GPI-anchored MT4-MMP. Here, we investigated the fate and cellular trafficking of MT4-MMP by analysing its homophilic complex interactions, internalization and recycling dynamics as compared with an inert form, MT4-MMP-E249A. Oligomeric and dimeric complexes were analysed by cotransfection of cells with FLAG-tagged or Myc-tagged MT4-MMP in reducing and nonreducing immunoblotting and coimmunoprecipitation experiments. The trafficking of MT4-MMP was studied with an antibody feeding assay and confocal microscopy analysis or cell surface protein biotinylation and western blot analysis. We demonstrate that MT4-MMP forms homophilic complexes at the cell surface, and internalizes in early endosomes, and that some of the enzyme is either autodegraded or recycled to the cell surface. Our data indicate that MT4-MMP is internalized by the clathrin-independent carriers/GPI-enriched early endosomal compartments pathway, a mechanism that differs from that responsible for the internalization of other membrane-type MMP members. Although MT4-MMP localizes with caveolin-1, MT4-MMP internalization was not affected by inhibitors of caveolin-1 or clathrin endocytosis pathways, but was reduced by CDC42 or RhoA silencing with small interfering RNA. We provide a new mechanistic insight into the regulatory mechanisms of MT4-MMP, which may have implications for the design of novel therapeutic strategies for metastatic breast cancer.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Metaloproteinases da Matriz Associadas à Membrana/metabolismo , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Feminino , Humanos , Cinética , Metaloproteinases da Matriz Associadas à Membrana/genética
6.
Cancer Res ; 74(23): 6758-70, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25320013

RESUMO

MT4-MMP (MMP-17) is a glycosylphosphatidyl inositol-anchored matrix metalloprotease expressed on the surface of cancer cells that promotes tumor growth and metastasis. In this report, we identify MT4-MMP as an important driver of cancer cell proliferation through CDK4 activation and retinoblastoma protein inactivation. We also determine a functional link between MT4-MMP and the growth factor receptor EGFR. Mechanistic experiments revealed direct association of MT4-MMP and its positive effects on EGFR phosphorylation in response to TGFα and EGF in cancer cells. Notably, the effects of MT4-MMP on proliferation and EGFR activation did not rely on metalloprotease activity. Clinically, MT4-MMP and EGFR expressions were correlated in human triple-negative breast cancer specimens. Altogether, our results identify MT4-MMP as a positive modifier of EGFR outside-in signaling that acts to cooperatively drive cancer cell proliferation.


Assuntos
Neoplasias da Mama/metabolismo , Membrana Celular/metabolismo , Receptores ErbB/metabolismo , Metaloproteinases da Matriz Associadas à Membrana/metabolismo , Transdução de Sinais/fisiologia , Animais , Células COS , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Chlorocebus aethiops , Quinase 4 Dependente de Ciclina/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Feminino , Células HeLa , Humanos , Camundongos , Proteína do Retinoblastoma/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo
7.
Cell Metab ; 20(2): 280-94, 2014 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-25017943

RESUMO

The molecular mechanisms responsible for the failure of antiangiogenic therapies and how tumors adapt to these therapies are unclear. Here, we applied transcriptomic, proteomic, and metabolomic approaches to preclinical models and provide evidence for tumor adaptation to vascular endothelial growth factor blockade through a metabolic shift toward carbohydrate and lipid metabolism in tumors. During sunitinib or sorafenib treatment, tumor growth was inhibited and tumors were hypoxic and glycolytic. In sharp contrast, treatment withdrawal led to tumor regrowth, angiogenesis restoration, moderate lactate production, and enhanced lipid synthesis. This metabolic shift was associated with a drastic increase in metastatic dissemination. Interestingly, pharmacological lipogenesis inhibition with orlistat or fatty acid synthase downregulation with shRNA inhibited tumor regrowth and metastases after sunitinib treatment withdrawal. Our data shed light on metabolic alterations that result in cancer adaptation to antiangiogenic treatments and identify key molecules involved in lipid metabolism as putative therapeutic targets.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Lipídeos/biossíntese , Neoplasias/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Progressão da Doença , Ácido Graxo Sintases/antagonistas & inibidores , Ácido Graxo Sintases/genética , Ácido Graxo Sintases/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Indóis/uso terapêutico , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Metástase Neoplásica , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Niacinamida/análogos & derivados , Niacinamida/uso terapêutico , Compostos de Fenilureia/uso terapêutico , Proteômica , Pirróis/uso terapêutico , Interferência de RNA , Sorafenibe , Sunitinibe , Transplante Heterólogo
8.
Front Pharmacol ; 2: 111, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21687519

RESUMO

The development of vascular system depends on the coordinated activity of a number of distinct families of molecules including growth factors and their receptors, cell adhesion molecules, extracellular matrix (ECM) molecules, and proteolytic enzymes. Matrix metalloproteases (MMPs) are a family of ECM degrading enzymes required for both physiological and pathological angiogenesis. Increasing evidence, point to a direct role of membrane type-MMPs (MT-MMPs) in vascular system stabilization, maturation, and leakage. Our understanding of the nature of MT-MMP interaction with extracellular and cell surface molecules and their multiple roles in vessel walls and perivascular stroma may provide new insights into mechanisms underlying vascular cell-ECM interactions and cell fate decisions in pathological conditions. Regulation of vascular leakage by MT-MMP interactions with the ECM could also lead to novel targeting opportunities for drug delivery in tumor. This review will shed lights on the emerging roles of MT1-MMP and MT4-MMP in vascular system alterations associated with cancer progression.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA