Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 41(20): 2833-2845, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35418692

RESUMO

Dysregulation of pericellular proteolysis is strongly implicated in cancer metastasis through alteration of cell invasion and the microenvironment. Matriptase-2 (MT-2) is a membrane-anchored serine protease which can suppress prostate cancer (PCa) cell invasion. In this study, we showed that MT-2 was down-regulated in PCa and could suppress PCa cell motility, tumor growth, and metastasis. Using microarray and biochemical analysis, we found that MT-2 shifted TGF-ß action towards its tumor suppressor function by repressing epithelial-to-mesenchymal transition (EMT) and promoting Smad2 phosphorylation and nuclear accumulation to upregulate two TGF-ß1 downstream effectors (p21 and PAI-1), culminating in hindrance of PCa cell motility and malignant growth. Mechanistically, MT-2 could dramatically up-regulate the expression of nuclear receptor NR4A3 via iron metabolism in PCa cells. MT-2-induced NR4A3 further coactivated Smad2 to activate p21 and PAI-1 expression. In addition, NR4A3 functioned as a suppressor of PCa and mediated MT-2 signaling to inhibit PCa tumorigenesis and metastasis. These results together indicate that NR4A3 sustains MT-2 signaling to suppress PCa cell invasion, tumor growth, and metastasis, and serves as a contextual factor for the TGF-ß/Smad2 signaling pathway in favor of tumor suppression via promoting p21 and PAI-1 expression.


Assuntos
Proteínas de Ligação a DNA , Proteínas de Membrana , Neoplasias da Próstata , Receptores de Esteroides , Receptores dos Hormônios Tireóideos , Serina Endopeptidases , Linhagem Celular Tumoral , Movimento Celular , Proteínas de Ligação a DNA/metabolismo , Transição Epitelial-Mesenquimal , Humanos , Masculino , Proteínas de Membrana/metabolismo , Invasividade Neoplásica , Inibidor 1 de Ativador de Plasminogênio , Próstata/patologia , Neoplasias da Próstata/patologia , Receptores de Esteroides/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo , Serina Endopeptidases/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Microambiente Tumoral
2.
Oncogene ; 39(37): 5950-5963, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32778768

RESUMO

TMPRSS2 is an important membrane-anchored serine protease involved in human prostate cancer progression and metastasis. A serine protease physiologically often comes together with a cognate inhibitor for execution of proteolytically biologic function; however, TMPRSS2's cognate inhibitor is still elusive. To identify the cognate inhibitor of TMPRSS2, in this study, we applied co-immunoprecipitation and LC/MS/MS analysis and isolated hepatocyte growth factor activator inhibitors (HAIs) to be potential inhibitor candidates for TMPRSS2. Moreover, the recombinant HAI-2 proteins exhibited a better inhibitory effect on TMPRSS2 proteolytic activity than HAI-1, and recombinant HAI-2 proteins had a high affinity to form a complex with TMPRSS2. The immunofluorescence images further showed that TMPRSS2 was co-localized to HAI-2. Both KD1 and KD2 domain of HAI-2 showed comparable inhibitory effects on TMPRSS2 proteolytic activity. In addition, HAI-2 overexpression could suppress the induction effect of TMPRSS2 on pro-HGF activation, extracellular matrix degradation and prostate cancer cell invasion. We further determined that the expression levels of TMPRSS2 were inversely correlated with HAI-2 levels during prostate cancer progression. In orthotopic xenograft animal model, TMPRSS2 overexpression promoted prostate cancer metastasis, and HAI-2 overexpression efficiently blocked TMPRSS2-induced metastasis. In summary, the results together indicate that HAI-2 can function as a cognate inhibitor for TMPRSS2 in human prostate cancer cells and may serve as a potential factor to suppress TMPRSS2-mediated malignancy.


Assuntos
Glicoproteínas de Membrana/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Serina Endopeptidases/metabolismo , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Xenoenxertos , Humanos , Masculino , Glicoproteínas de Membrana/química , Invasividade Neoplásica , Neoplasias da Próstata/etiologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Proteínas Secretadas Inibidoras de Proteinases/metabolismo , Proteólise
4.
Oncogene ; 38(28): 5580-5598, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31152147

RESUMO

Gram-negative bacteria have been found to be a major population in prostatitis and prostate cancer (PCa) tissues. Lipopolysaccharide (LPS), a major compound of Gram-negative bacteria, with stimulatory activities in some cancer types, but has not been fully studied in PCa. In this study, we examined the effect of LPS on the invasion of PCa cells. Interestingly, LPS can enhance the invasiveness of PCa, but had no significant effect on PCa cell viability. Using protease inhibitor screening and biochemical analyses, matriptase, a member of the membrane-anchored serine protease family, is found to play a key role in LPS-induced PCa cell invasion. Mechanistically, Toll-like receptor 4 (TLR4, LPS receptor)-sphingosine kinase 1 (SphK1) signaling underlies LPS-induced matriptase activation and PCa cell invasion. Specifically, LPS induced the S225 phosphorylation of SphK1 and the translocation of SphK1 to plasma membrane, leading to the production of sphingosine 1-phosphate (S1P), ERK1/2 and matriptase activation via S1P receptor 4 (S1PR4). This phenomenon is further validated using the patient-derived explant (PDE) model. Indeed, there is a significant correlation among the elevated SphK1 levels, the Gleason grades of PCa specimens, and the poor survival of PCa patients. Taken together, this study demonstrates a potential impact of LPS on PCa progression. Our results provide not only a new finding of the role of bacterial infection in PCa progression but also potential therapeutic target(s) associated with PCa metastasis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Invasividade Neoplásica , Metástase Neoplásica , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Polissacarídeos/farmacologia , Neoplasias da Próstata/patologia , Serina Endopeptidases/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Progressão da Doença , Ativação Enzimática , Humanos , Masculino , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/metabolismo
5.
Br J Cancer ; 120(5): 499-511, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30765871

RESUMO

BACKGROUND: Dysregulation of pericellular proteolysis usually accounts for cancer cell invasion and metastasis. Isolation of a cell-surface protease system for lung cancer metastasis is an important issue for mechanistic studies and therapeutic target identification. METHODS: Immunohistochemistry of a tissue array (n = 64) and TCGA database (n = 255) were employed to assess the correlation between serine protease inhibitors (SPIs) and lung adenocarcinoma progression. The role of SPI in cell motility was examined using transwell assays. Pulldown and LC/MS/MS were performed to identify the SPI-modulated novel protease(s). A xenografted mouse model was harnessed to demonstrate the role of the SPI in lung cancer metastasis. RESULTS: Hepatocyte growth factor activator inhibitor-2 (HAI-2) was identified to be downregulated following lung cancer progression, which was related to poor survival and tumour invasion. We further isolated a serum-derived serine protease, plasmin, to be a novel target of HAI-2. Downregulation of HAI-2 promotes cell surface plasmin activity, EMT, and cell motility. HAI-2 can suppress plasmin-mediated activations of HGF and TGF-ß1, EMT and cell invasion. In addition, downregulated HAI-2 increased metastasis of lung adenocarcinoma via upregulating plasmin activity. CONCLUSION: HAI-2 functions as a novel inhibitor of plasmin to suppress lung cancer cell motility, EMT and metastasis.


Assuntos
Adenocarcinoma de Pulmão/metabolismo , Fibrinolisina/metabolismo , Neoplasias Pulmonares/metabolismo , Glicoproteínas de Membrana/metabolismo , Células A549 , Adenocarcinoma de Pulmão/patologia , Adenocarcinoma de Pulmão/secundário , Animais , Linhagem Celular Tumoral , Movimento Celular , Progressão da Doença , Transição Epitelial-Mesenquimal , Fibrinolisina/antagonistas & inibidores , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Fator de Crescimento Transformador beta1/metabolismo
6.
Sci Rep ; 7(1): 15101, 2017 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-29118397

RESUMO

Dysregulation of pericellular proteolysis is often required for tumor invasion and cancer progression. It has been shown that down-regulation of hepatocyte growth factor activator inhibitor-2 (HAI-2) results in activation of matriptase (a membrane-anchored serine protease), human prostate cancer cell motility and tumor growth. In this study, we further characterized if HAI-2 was a cognate inhibitor for matriptase and identified which Kunitz domain of HAI-2 was required for inhibiting matriptase and human prostate cancer cell motility. Our results show that HAI-2 overexpression suppressed matriptase-induced prostate cancer cell motility. We demonstrate that HAI-2 interacts with matriptase on cell surface and inhibits matriptase proteolytic activity. Moreover, cellular HAI-2 harnesses its Kunitz domain 1 (KD1) to inhibit matriptase activation and prostate cancer cell motility although recombinant KD1 and KD2 of HAI-2 both show an inhibitory activity and interaction with matriptase protease domain. The results together indicate that HAI-2 is a cognate inhibitor of matriptase, and KD1 of HAI-2 plays a major role in the inhibition of cellular matritptase activation as well as human prostate cancer invasion.


Assuntos
Movimento Celular , Glicoproteínas de Membrana/metabolismo , Domínios Proteicos , Serina Endopeptidases/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Células HEK293 , Humanos , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Camundongos Endogâmicos BALB C , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteólise , Interferência de RNA , Homologia de Sequência de Aminoácidos , Serina Endopeptidases/genética , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/genética , Inibidores de Serina Proteinase/metabolismo
7.
Cancer Res ; 75(14): 2949-60, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-26018085

RESUMO

Dysregulation of androgen signaling and pericellular proteolysis is necessary for prostate cancer progression, but the links between them are still obscure. In this study, we show how the membrane-anchored serine protease TMPRSS2 stimulates a proteolytic cascade that mediates androgen-induced prostate cancer cell invasion, tumor growth, and metastasis. We found that matriptase serves as a substrate for TMPRSS2 in mediating this proinvasive action of androgens in prostate cancer. Further, we determined that higher levels of TMPRSS2 expression correlate with higher levels of matriptase activation in prostate cancer tissues. Lastly, we found that the ability of TMPRSS2 to promote prostate cancer tumor growth and metastasis was associated with increased matriptase activation and enhanced degradation of extracellular matrix nidogen-1 and laminin ß1 in tumor xenografts. In summary, our results establish that TMPRSS2 promotes the growth, invasion, and metastasis of prostate cancer cells via matriptase activation and extracellular matrix disruption, with implications to target these two proteases as a strategy to treat prostate cancer.


Assuntos
Androgênios/farmacologia , Matriz Extracelular/metabolismo , Neoplasias da Próstata/patologia , Serina Endopeptidases/metabolismo , Serina Endopeptidases/fisiologia , Animais , Células CHO , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Cricetinae , Cricetulus , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Matriz Extracelular/patologia , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Nus , Camundongos SCID , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Serina Endopeptidases/genética , Células Tumorais Cultivadas , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
8.
Am J Pathol ; 177(6): 3145-58, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20971737

RESUMO

Deregulation of both ErbB-2 signaling and matriptase activity has been associated with human prostate cancer (PCa) progression. In this communication, we investigated the roles of both ErbB-2 signaling in matriptase zymogen activation and matriptase in ErbB-2-induced PCa malignancy. In a human PCa cell progression model, we observed that advanced PCa C-81 LNCaP cells exhibited an aggressive phenotype with increased cell migration and invasion capacity; these cells concurrently showed both enhanced ErbB-2 phosphorylation and increased matriptase zymogen activation compared with parental C-33 LNCaP cells. Moreover, ErbB2 activation, both ligand-dependent (eg, epidermal growth factor treatment) and ligand-independent (eg, overexpression), was able to induce matriptase zymogen activation in this cell line. Inhibition of ErbB-2 activity by either the specific inhibitor, AG825, in epidermal growth factor-treated C-33 LNCaP cells or ErbB-2 knockdown in C-81 LNCaP cells, reduced matriptase activation. These observations were confirmed by similar studies using both DU145 and PC3 cells. Together, these data suggest that ErbB-2 signaling plays an important role in matriptase zymogen activation. ErbB-2-enhanced matriptase activation was suppressed by a phosphatidylinositol 3-kinase inhibitor (ie, LY294002) but not by a MEK inhibitor (ie, PD98059). Suppression of matriptase expression by small hairpin RNA knockdown in ErbB-2-overexpressing LNCaP cells dramatically suppressed cancer cell invasion. In summary, our data indicate that ErbB-2 signaling via the phosphatidylinositol 3-kinase pathway results in up-regulated matriptase zymogen activity, which contributes to PCa cell invasion.


Assuntos
Carcinoma/patologia , Genes erbB-2/fisiologia , Neoplasias da Próstata/patologia , Serina Endopeptidases/fisiologia , Androgênios/farmacologia , Carcinoma/genética , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Células Cultivadas , Progressão da Doença , Resistência a Medicamentos/efeitos dos fármacos , Resistência a Medicamentos/genética , Ativação Enzimática/genética , Técnicas de Silenciamento de Genes , Humanos , Masculino , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Neoplasias da Próstata/genética , RNA Interferente Pequeno/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA