Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Respir Res ; 17: 17, 2016 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-26887531

RESUMO

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a chronic progressively fatal disease. Extracellular Matrix Metalloproteinase Inducer (EMMPRIN) is a glycosylated transmembrane protein that induces the expression of some matrix metalloproteinase (MMP) in neighboring stromal cells through direct epithelial-stromal interactions. EMMPRIN is highly expressed in type II alveolar epithelial cells at the edges of the fibrotic areas in IPF lung sections. However, the exact role of EMMPRIN in IPF is unknown. METHODS: To determine if EMMPRIN contributes to lung fibroblast proliferation, resistance to apoptosis, and differentiation to myofibroblasts, normal Human lung fibroblasts (NHLF) transiently transfected with either EMMPRIN/GFP or GFP were treated with TGF- ß1 from 0 to 10 ng/ml for 48 h and examined for cell proliferation (thymidine incorporation), apoptosis (FACS analysis and Cell Death Detection ELISA assay), cell migration (Modified Boyden chamber) and differentiation to myofibroblasts using Western blot for α-smooth actin of cell lysates. The effect of EMMPRIN inhibition on NHLF proliferation, apoptosis, migration and differentiation to myofibroblasts after TGF- ß1 treatment was examined using EMMPRIN blocking antibody. We examined the mechanism by which EMMPRIN induces its effects on fibroblasts by studying the ß-catenin/canonical Wnt signaling pathway using Wnt luciferase reporter assays and Western blot for total and phosphorylated ß-catenin. RESULTS: Human lung fibroblasts overexpressing EMMPRIN had a significant increase in cell proliferation and migration compared to control fibroblasts. Furthermore, EMMPRIN promoted lung fibroblasts resistance to apoptosis. Lung fibroblasts overexpressing EMMPRIN showed a significantly increased expression of α- smooth muscle actin, a marker of differentiation to myofibroblasts compared to control cells. TGF-ß1 increased the expression of EMMPRIN in lung fibroblasts in a dose-dependent manner. Attenuation of EMMPRIN expression with the use of an EMMPRIN blocking antibody markedly inhibited TGF-ß1 induced proliferation, migration, and differentiation of fibroblasts to myofibroblasts. EMMPRIN overexpression in lung fibroblasts was found to induce an increase in TOPFLASH luciferase reporter activity when compared with control fibroblasts. CONCLUSION: These findings indicate that TGF-ß1 induces the release of EMMPRIN that activates ß-catenin/canonical Wnt signaling pathway. EMMPRIN overexpression induces an anti-apoptotic and pro-fibrotic phenotype in lung fibroblasts that may contribute to the persistent fibro-proliferative state seen in IPF.


Assuntos
Basigina/metabolismo , Fibroblastos/citologia , Fibroblastos/fisiologia , Pulmão/citologia , Pulmão/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Humanos , Miofibroblastos/citologia , Miofibroblastos/fisiologia
2.
Mol Pharmacol ; 87(3): 501-12, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25552486

RESUMO

Because cancer cell invasion is a critical determinant of metastasis, targeting invasion is a viable approach to prevent metastasis. Utilizing a novel three-dimensional high-throughput invasion assay, we screened a National Cancer Institute compound library and discovered compounds demonstrating inhibitory effects on cancer cell invasion. One hit, trifluoperazine, suppresses invasion of human cancer cell lines while displaying a limited cytotoxicity profile. This inhibition is due to the interference with cancer cell migratory ability but not proteolytic activity. Treatment of cancer cells with trifluoperazine significantly reduces angiogenesis and prevents cancer cell invasion through a chorioallantoic basement membrane. Mechanistically, treatment results in decreased phosphorylated AKT (Ser(473) and Thr(308)) and ß-catenin (Ser(552)). Lack of phosphorylation of Ser(552) of ß-catenin prevents ß-catenin nuclear relocation, resulting in decreased expression of vascular endothelial growth factor, likely mediated through dopamine receptor D2. Taken together, we demonstrated that trifluoperazine is responsible for reducing the angiogenic and invasive potential of aggressive cancer cells through dopamine receptor D2 to modulate the ß-catenin pathway and propose that trifluoperazine may be used as an antimetastasis chemotherapeutic.


Assuntos
Antineoplásicos/farmacologia , Antipsicóticos/farmacologia , Invasividade Neoplásica/prevenção & controle , Trifluoperazina/farmacologia , Animais , Linhagem Celular Tumoral , Embrião de Galinha , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Células NIH 3T3 , Invasividade Neoplásica/patologia
3.
J Biol Chem ; 286(38): 33167-77, 2011 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-21795678

RESUMO

Matrix metalloproteinases (MMPs) have been shown to be key players in both extracellular matrix remodeling and cell migration during cancer metastasis. MMP-14, a membrane-anchored MMP, in particular, is closely associated with these processes. The hemopexin (PEX) domain of MMP-14 has been proposed as the modulating region involved in the molecular cross-talk that initiates cell migration through homodimerization of MMP-14 as well as heterodimerization with the cell surface adhesion molecule CD44. In this study, minimal regions required for function within the PEX domain were investigated through a series of substitution mutations. Blades I and IV were found to be involved in cell migration. We found that blade IV is necessary for MMP-14 homodimerization and that blade I is required for CD44 MMP-14 heterodimerization. Cross-talk between MMP-14 and CD44 results in phosphorylation of EGF receptor and downstream activation of the MAPK and PI3K signaling pathways involved in cell migration. Based on these mutagenesis analyses, peptides mimicking the essential outermost strand motifs within the PEX domain of MMP-14 were designed. These synthetic peptides inhibit MMP-14-enhanced cell migration in a dose-dependent manner but have no effect on the function of other MMPs. Furthermore, these peptides interfere with cancer metastasis without affecting primary tumor growth. Thus, targeting the MMP-14 hemopexin domain represents a novel approach to inhibit MMP-14-mediated cancer dissemination.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Movimento Celular , Inibidores de Metaloproteinases de Matriz , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/mortalidade , Células COS , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Galinhas , Chlorocebus aethiops , Ativação Enzimática/efeitos dos fármacos , Receptores ErbB/metabolismo , Feminino , Humanos , Receptores de Hialuronatos/metabolismo , Metaloproteinase 14 da Matriz/química , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Dados de Sequência Molecular , Neovascularização Patológica/enzimologia , Peptídeos/química , Peptídeos/farmacologia , Estrutura Terciária de Proteína , Transdução de Sinais/efeitos dos fármacos , Taxa de Sobrevida
4.
Methods Mol Biol ; 1406: 195-202, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26820957

RESUMO

In order to more rapidly define the mechanism by which certain drugs and compounds can influence cancer cell invasion, we have combined a traditional phagokinetic gold migration assay with a fluorescent substrate. The purpose of this dual assay is to provide a platform by which to simultaneously monitor proteolytic activity and cancer cell migratory ability, both of which are required for the crucial step of cancer cell invasion during metastasis. This assay allows for delineation of potential mechanisms of action a compound of interest has, as one can determine whether or not a cancer cell that is being treated with the potential drug has changes in proteolytic activity and/or migratory ability at the same time.


Assuntos
Ensaios de Migração Celular/métodos , Corantes Fluorescentes/metabolismo , Fagocitose , Linhagem Celular , Gelatina/química , Ouro/química , Proteólise , Espectrometria de Fluorescência , Fatores de Tempo , Xantenos/química
5.
Front Biosci (Schol Ed) ; 7(1): 125-49, 2015 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-25961691

RESUMO

Matrix metalloproteinases (MMPs) were identified as early as 1962. Since this seminal finding, this family of zinc-dependent endopeptidases has been studied extensively. This collective work has resulted in delineation of MMP gene and protein structures, the mechanisms of control of MMPs, the action of MMPs on both extracellular matrices and other proteins such as growth factors and cytokines, naturally-occurring mechanisms of control, and of course their role in normal physiology and their crucial roles in pathophysiology. Stemming from the discovery that MMPs contribute to arthritis, heart disease, and cancer, amongst other diseases, attempts to develop treatment strategies incorporating MMP inhibition have been undertaken. The results of these endeavours have been mediocre, resulting in few FDA-approved MMP inhibitors mostly due to the broad-spectrum nature of these early inhibitors and unwanted side effects of MMP inhibition. The future of exploitation of MMPs in disease lies in the design of more targeted inhibitors; in order to accomplish this, we must all understand the subtle differences between each MMP and their contextual roles. In this chapter, we aim to overview major topics regarding MMPs and what direction we may go in the future.


Assuntos
Metaloproteinases da Matriz/química , Metaloproteinases da Matriz/metabolismo , Animais , Humanos , Metaloproteinases da Matriz/genética
6.
Genes Dis ; 2(`1): 26-34, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26097889

RESUMO

Since the identification of matrix metalloproteinases (MMPs), a family of zinc-dependent endopeptidases, as being a driving factor for cancer progression and patient prognosis, MMPs have been studied extensively. Although early programs targeting MMPs were largely unsuccessful in clinical trials, they remain a viable and highly desirable therapeutic target based on preclinical studies and their role in disease progression. As information regarding the structure and function of these proteinases is compiled and biotechnology evolves, tools to develop better inhibitors is within our grasp. Improved methods for high throughput screening and in silico drug design programs have identified compounds which are highly potent, have high binding affinities, and exhibit favorable pharmacokinetic profiles. More recently, advances in drug delivery methods or compounds which bind outside the active site have brought new light to the field. In this review, we highlight the role of MMPs in cancer, clinical trials for MMP inhibitors, and novel approaches to targeting MMPs in cancer.

7.
Cancer Res ; 75(13): 2674-85, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25977338

RESUMO

Upregulation of matrix metalloproteinase MMP-14 (MT1-MMP) is associated with poor prognosis in cancer patients, but it is unclear how MMP-14 becomes elevated in tumors. Here, we show that miR-181a-5p is downregulated in aggressive human breast and colon cancers where its levels correlate inversely with MMP-14 expression. In clinical specimens, enhanced expression of MMP-14 was observed in cancer cells located at the invasive front of tumors where miR-181a-5p was downregulated relative to adjacent normal cells. Bioinformatics analyses defined a potential miR-181a-5p response element within the 3'-untranslated region of MMP-14 that was validated in reporter gene experiments. Ectopic miR-181a-5p reduced MMP-14 expression, whereas miR-181a-5p attenuation elevated MMP-14 expression. In support of a critical relationship between these two genes, miR-181a-5p-mediated reduction of MMP-14 levels was sufficient to decrease cancer cell migration, invasion, and activation of pro-MMP-2. Furthermore, this reduction in MMP-14 levels was sufficient to reduce in vivo invasion and angiogenesis in chick chorioallantoic membrane assays. Taken together, our results establish the regulation of MMP-14 in cancers by miR-181a-5p through a posttranscriptional mechanism, and they further suggest strategies to elevate miR-181a-5p to prevent cancer metastasis.


Assuntos
Neoplasias da Mama/metabolismo , Movimento Celular/fisiologia , Neoplasias do Colo/metabolismo , Metaloproteinase 14 da Matriz/biossíntese , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias do Colo/irrigação sanguínea , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Regulação para Baixo , Feminino , Humanos , Metaloproteinase 14 da Matriz/genética , MicroRNAs/genética , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Elementos de Resposta , Transfecção , Regulação para Cima
8.
J Natl Cancer Inst ; 105(18): 1402-16, 2013 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-23990668

RESUMO

BACKGROUND: Cell migration is a critical determinant of cancer metastasis, and a better understanding of the genes involved will lead to the identification of novel targets aimed at preventing cancer dissemination. KIAA1199 has been shown to be upregulated in human cancers, yet its role in cancer progression was hitherto unknown. METHODS: Clinical relevance was assessed by examining KIAA1199 expression in human cancer specimens. In vitro and in vivo studies were employed to determine the function of KIAA1199 in cancer progression. Cellular localization of KIAA1199 was microscopically determined. SNAP-tag pull-down assays were used to identify binding partner(s) of KIAA1199. Calcium levels were evaluated using spectrofluorometric and fluorescence resonance energy transfer analyses. Signaling pathways were dissected by Western blotting. Student t test was used to assess differences. All statistical tests were two-sided. RESULTS: KIAA1199 was upregulated in invasive breast cancer specimens and inversely associated with patient survival rate. Silencing of KIAA1199 in MDA-MB-435 cancer cells resulted in a mesenchymal-to-epithelial transition that reduced cell migratory ability in vitro (75% reduction; P < .001) and decreased metastasis in vivo (80% reduction; P < .001). Gain-of-function assays further demonstrated the role of KIAA1199 in cell migration. KIAA1199-enhanced cell migration required endoplasmic reticulum (ER) localization, where it forms a stable complex with the chaperone binding immunoglobulin protein (BiP). A novel ER-retention motif within KIAA1199 that is required for its ER localization, BiP interaction, and enhanced cell migration was identified. Mechanistically, KIAA1199 was found to mediate ER calcium leakage, and the resultant increase in cytosolic calcium ultimately led to protein kinase C alpha activation and cell migration. CONCLUSIONS: KIAA1199 serves as a novel cell migration-promoting gene and plays a critical role in maintaining cancer mesenchymal status.


Assuntos
Cálcio/metabolismo , Movimento Celular , Retículo Endoplasmático/metabolismo , Inativação Gênica , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas/metabolismo , Animais , Western Blotting , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Progressão da Doença , Transição Epitelial-Mesenquimal , Recuperação de Fluorescência Após Fotodegradação , Transferência Ressonante de Energia de Fluorescência , Regulação Neoplásica da Expressão Gênica , Humanos , Hialuronoglucosaminidase , Camundongos , Metástase Neoplásica , Transplante de Neoplasias , Proteína Quinase C-alfa/metabolismo , Proteínas/genética , Transdução de Sinais , Espectrometria de Fluorescência , Regulação para Cima
9.
PLoS One ; 7(6): e38403, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22679501

RESUMO

Emerging evidence has implicated the role of tumor initiating cells (TICs) in the process of cancer metastasis. The mechanism underlying the conversion of TICs from stationary to invasive remains to be characterized. In this report, we employed less invasive breast cancer TICs, SK-3rd, that displays CD44(high)/CD24(low) with high mammosphere-forming and tumorigenic capacities, to investigate the mechanism by which stationary TICs are converted to invasive TICs. Invasive ability of SK-3rd TICs was markedly enhanced when the cells were cultured under hypoxic conditions. Given the role of membrane type 1-matrix metalloproteinase (MT1-MMP) in cancer invasion/metastasis, we explored a possible involvement of MT1-MMP in hypoxia-induced TIC invasion. Silencing of MT1-MMP by a shRNA approach resulted in diminution of hypoxia-induced cell invasion in vitro and metastasis in vivo. Under hypoxic conditions, MT1-MMP redistributed from cytoplasmic storage pools to the cell surface of TICs, which coincides with the increased cell invasion. In addition, CD44, a cancer stem-like cell marker, inversely correlated with increased cell surface MT1-MMP. Interestingly, cell surface MT1-MMP gradually disappeared when the hypoxia-treated cells were switched to normoxia, suggesting the plasticity of TICs in response to oxygen content. Furthermore, we dissected the pathways leading to upregulated MT1-MMP in cytoplasmic storage pools under normoxic conditions, by demonstrating a cascade involving Twist1-miR10b-HoxD10 leading to enhanced MT1-MMP expression in SK-3rd TICs. These observations suggest that MT1-MMP is a key molecule capable of executing conversion of stationary TICs to invasive TICs under hypoxic conditions and thereby controlling metastasis.


Assuntos
Neoplasias da Mama/metabolismo , Hipóxia Celular/fisiologia , Metaloproteinase 14 da Matriz/metabolismo , Transporte Proteico/fisiologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Transporte Proteico/genética , Reação em Cadeia da Polimerase em Tempo Real
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa