Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cancer ; 22(1): 4, 2023 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-36624516

RESUMO

BACKGROUND: Metastatic colonization is one of the critical steps in tumor metastasis. A pre-metastatic niche is required for metastatic colonization and is determined by tumor-stroma interactions, yet the mechanistic underpinnings remain incompletely understood. METHODS: PCR-based miRNome profiling, qPCR, immunofluorescent analyses evaluated the expression of exosomal miR-141 and cell-to-cell communication. LC-MS/MS proteomic profiling and Dual-Luciferase analyses identified YAP1 as the direct target of miR-141. Human cytokine profiling, ChIP, luciferase reporter assays, and subcellular fractionation analyses confirmed YAP1 in modulating GROα production. A series of in vitro tumorigenic assays, an ex vivo model and Yap1 stromal conditional knockout (cKO) mouse model demonstrated the roles of miR-141/YAP1/GROα/CXCR1/2 signaling cascade. RNAi, CRISPR/Cas9 and CRISPRi systems were used for gene silencing. Blood sera, OvCa tumor tissue samples, and tissue array were included for clinical correlations. RESULTS: Hsa-miR-141-3p (miR-141), an exosomal miRNA, is highly secreted by ovarian cancer cells and reprograms stromal fibroblasts into proinflammatory cancer-associated fibroblasts (CAFs), facilitating metastatic colonization. A mechanistic study showed that miR-141 targeted YAP1, a critical effector of the Hippo pathway, reducing the nuclear YAP1/TAZ ratio and enhancing GROα production from stromal fibroblasts. Stromal-specific knockout (cKO) of Yap1 in murine models shaped the GROα-enriched microenvironment, facilitating in vivo tumor colonization, but this effect was reversed after Cxcr1/2 depletion in OvCa cells. The YAP1/GROα correlation was demonstrated in clinical samples, highlighting the clinical relevance of this research and providing a potential therapeutic intervention for impeding premetastatic niche formation and metastatic progression of ovarian cancers. CONCLUSIONS: This study uncovers miR-141 as an OvCa-derived exosomal microRNA mediating the tumor-stroma interactions and the formation of tumor-promoting stromal niche through activating YAP1/GROα/CXCRs signaling cascade, providing new insight into therapy for OvCa patients with peritoneal metastases.


Assuntos
MicroRNAs , Neoplasias Ovarianas , Humanos , Animais , Camundongos , Feminino , Cromatografia Líquida , Proteômica , Espectrometria de Massas em Tandem , Neoplasias Ovarianas/genética , MicroRNAs/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Microambiente Tumoral
2.
Mol Cancer ; 16(1): 11, 2017 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-28095864

RESUMO

BACKGROUND: Cancer metastasis is determined by the formation of the metastatic niche and the ability of cancer cells to adapt to microenvironmental stresses. Anoikis resistance is a fundamental feature of metastatic cancer cell survival during metastatic cancer progression. However, the mechanisms underlying anoikis resistance in ovarian cancer are still unclear. METHODS: Expressions of miRNA-141 and its downstream targets were evaluated by qPCR, Western blotting, Immunohistochemical (IHC) and in situ hybridization (ISH) assays. The luciferase assays were used to prove KLF12 as the downstream target of miR-141. The cDNA microarray and apoptotic protein arrays were used to identify the targets of miR-141 and KLF12. The competition of KLF12 and Sp1 on survivin promoter was examined by ChIP assay. IHC analysis on ovarian cancer tissue array was used to evaluate the expressions of KLF12 and miR-141 and to show the clinical relevance. The functional studies were performed by in vitro and in vivo tumorigenic assays. RESULTS: Enforced expression of miR-141 promotes, while knockdown of miR-141 expression inhibits, cell proliferation, anchorage-independent capacity, anoikis resistance, tumor growth and peritoneal metastases of ovarian cancer cells. Bioinformatics and functional analysis identified that Kruppel-related zinc finger protein AP-2rep (KLF12) is directly targeted by miR-141. Consistent with this finding, knockdown of KLF12 phenocopied the effects of miR-141 overexpression in ovarian cancer cells. In contrast, restoration of KLF12 in miR-141-expressing cells significantly attenuated anoikis resistance in ovarian cancer cells via interfering with Sp1-mediated survivin transcription, which inhibits the intrinsic apoptotic pathway and is crucial for ovarian cancer cell survival, anoikis resistance and peritoneal metastases. Immunohistochemical (IHC) and in situ hybridization (ISH) assays confirmed that miRNA-141 expression is inversely correlated with KLF12 expression and significantly associated with advanced ovarian cancers accompanied with distal metastases, underscoring the clinical relevance of our findings. CONCLUSIONS: Our data identify a novel signaling axis of miR-141/KLF12/Sp1/survivin in enhancing anoikis resistance and likely serves as a potential therapeutic target for metastatic ovarian cancer.


Assuntos
Anoikis/genética , Proteínas Inibidoras de Apoptose/genética , Fatores de Transcrição Kruppel-Like/genética , MicroRNAs/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Fator de Transcrição Sp1/genética , Animais , Sítios de Ligação , Movimento Celular/genética , Proliferação de Células , Sobrevivência Celular/genética , Modelos Animais de Doenças , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Metástase Neoplásica , Interferência de RNA , RNA Mensageiro/genética , Survivina , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Cell Rep ; 21(10): 2785-2795, 2017 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-29212026

RESUMO

Angiogenesis inhibitors are important for cancer therapy, but clinically approved anti-angiogenic agents have shown only modest efficacy and can compromise wound healing. This necessitates the development of novel anti-angiogenesis therapies. Here, we show significantly increased EGFL6 expression in tumor versus wound or normal endothelial cells. Using a series of in vitro and in vivo studies with orthotopic and genetically engineered mouse models, we demonstrate the mechanisms by which EGFL6 stimulates tumor angiogenesis. In contrast to its antagonistic effects on tumor angiogenesis, EGFL6 blockage did not affect normal wound healing. These findings have significant implications for development of anti-angiogenesis therapies.


Assuntos
Glicoproteínas/metabolismo , Proteínas de Neoplasias/metabolismo , Peptídeos/metabolismo , Animais , Western Blotting , Proteínas de Ligação ao Cálcio , Moléculas de Adesão Celular , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/fisiologia , Quitosana/metabolismo , Feminino , Glicoproteínas/genética , Humanos , Técnicas In Vitro , Integrinas/genética , Integrinas/metabolismo , Camundongos , Camundongos Knockout , Nanopartículas/química , Proteínas de Neoplasias/genética , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Peptídeos/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/genética , Fosforilação/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Proteína 1 Relacionada a Twist/genética , Proteína 1 Relacionada a Twist/metabolismo , Cicatrização/genética , Cicatrização/fisiologia
4.
PLoS One ; 8(1): e53597, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23301094

RESUMO

Recent evidence has suggested that AMPK activators may be applied as therapeutic drugs in suppressing cancer cell growth. However, the molecular mechanism of their suppressive function in cancer cells is still unclear. Here we show that AMPK activators impair cervical cancer cell growth through the reduction of DVL3, a positive regulator in Wnt/ß-catenin signaling and an oncogenic player in cervical cancer tumorigenesis. By western blot and immunohistochemical analyses, we demonstrated that DVL3 was frequently upregulated and significantly associated with elevated ß-catenin (P = 0.009) and CyclinD1 (P = 0.009) expressions in cervical cancer. Enforced expression of DVL3 elevated ß-catenin and augmented cervical cancer cell growth, verifying that DVL3-mediated Wnt/ß-catenin activation is involved in cervical cancer oncogenesis. On the other aspect, we noted that the cervical cancer cell growth was remarkably suppressed by AMPK activators and such cell growth inhibition was in concomitant with the reduction of DVL3 protein level in dose- and time-dependent manners. Besides, impaired mTOR signaling activity also reduced DVL3 expression. In contrast, co-treatment with Compound C (AMPK inhibitor) could significantly abrogate metformin induced DVL3 reduction. In addition, co-treatment with AM114 or MG132 (proteosomal inhibitors) could partially restore DVL3 expression under the treatment of metformin. Further in vivo ubiquitination assay revealed that metformin could reduce DVL3 by ubiquitin/proteasomal degradation. To our knowledge, this is the first report showing the probable molecular mechanisms of that the AMPK activators suppress cervical cancer cell growth by impairing DVL3 protein synthesis via AMPK/mTOR signaling and/or partially promoting the proteasomal degradation of DVL3.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fosfoproteínas/metabolismo , Neoplasias do Colo do Útero/enzimologia , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Proteínas Desgrenhadas , Relação Dose-Resposta a Droga , Ativação Enzimática , Feminino , Células HEK293 , Células HeLa , Humanos , Imuno-Histoquímica/métodos , Plasmídeos/metabolismo , Transdução de Sinais , Fatores de Tempo , Neoplasias do Colo do Útero/metabolismo
5.
Oncotarget ; 3(12): 1546-56, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23295859

RESUMO

Although the mortality rate of endometrial cancer is comparatively low in gynecologic malignancies, a rising trend of this cancer has been observed for the past decade. The understanding of the molecular mechanism will favor for the clinical management of this disease. Aberrant activation of Wnt/ß-catenin signaling pathway plays a major role in the pathogenesis of endometrioid adenocarcinoma including this cancer type. In this study, we reported that Sox7, one of Sox transcriptional factors, was frequently underexpressed in endometrial cancer and importantly, it was associated with dysregulation of the Wnt/ß-catenin signaling activity. Immunohistochemical and quantitative RT-PCR analyses showed that Sox7 was underexpressed and was associated with high-grade tumor (P=0.021), increased expressions of ß-catenin (P=0.038) and its downstream targets; CyclinD1 (P less than 0.001) and FGF9 (P less than 0.001). In addition, using HEK293T cell model, we found that Sox7 was able to inhibit TCF/LEF-1-dependent luciferase activity induced by Wnt-1. This was further proved by that Sox7 could significantly suppress the expressions of Wnt targets; Cyclin D1 and C-myc in endometrial cells. Immuno-fluorescent microscopy revealed that Sox7 was co-localizaed with either mutant ß-catenin or TCF4 protein in nucleus, while co-immunopreciptation assay demonstrated that Sox7 could physically interact with not only wild-type but also mutant ß-catenin, as well as TCF4 proteins. Functionally, enforced expression of Sox7 could significantly inhibit endometrial or endometrioid ovarian cancer cells (OEA) harboring either wild-type or mutant ß-catenin. These data suggest Sox7 is a negative regulator of Wnt/ß-catenin signaling pathway through impeding the transcriptional machinery of ß-catenin/TCF/LEF-1 transcriptional complex, and the loss of expression may be involved in the pathogenesis of endometrial cancer.


Assuntos
Neoplasias do Endométrio/metabolismo , Fatores de Transcrição SOXF/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Estudos de Casos e Controles , Linhagem Celular Tumoral , Sobrevivência Celular , Ciclina D1/metabolismo , Regulação para Baixo , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Feminino , Fator 9 de Crescimento de Fibroblastos/metabolismo , Genes Reporter , Células HEK293 , Humanos , Imuno-Histoquímica , Imunoprecipitação , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Microscopia de Fluorescência , Mutação , Gradação de Tumores , Estadiamento de Neoplasias , Proteínas Proto-Oncogênicas c-myc/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXF/genética , Fatores de Tempo , Fator de Transcrição 4 , Fatores de Transcrição/metabolismo , Transfecção , Proteínas Wnt/genética , Via de Sinalização Wnt/genética , Proteína Wnt1/metabolismo , Proteína Wnt3A/metabolismo , beta Catenina/genética
6.
PLoS One ; 7(12): e52578, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23285101

RESUMO

Ovarian cancer is a highly lethal disease with poor prognosis and especially in high-grade tumor. Emerging evidence has reported that aberrant upregulation and activation of GRB7, ERK as well as FOXM1 are closely associated with aggresivenesss of human cancers. However, the interplay between these factors in the pathogenesis of human cancers still remains unclear. In this study, we found that GRB7 (P<0.0001), ERK phosphorylation (P<0.0001) and FOXM1 (P = 0.001) were frequently increased and associated with high-grade tumors, as well as a high tendency in association with advanced stage ovarian cancer by immunohistochemical analysis. Intriguingly, the expressions of GRB7 (P<0.0001), ERK phosphorylation (P<0.001) and FOXM1 (P<0.001) showed a significant stepwise increase pattern along Grade 1 to Grade 3 ovarian cancers. Biochemical studies using western blot analysis demonstrated that enforced expression or knockdown of GRB7 showed GRB7 could elevate the levels of ERK phosphorylation and FOXM1, whereas enforced expression of FOXM1 could not alter levels of GRB7 and ERK phosphorylation. But inhibition of ERK signaling by U0126 or PD98059 could reduce the level of FOXM1 in GRB7-overexpressing ovarian cancer cells, suggesting that GRB7, ERK and FOXM1 are regulated orderly. Moreover, inhibition of ERK activity by U0126 or PD98059, or decreased FOXM1 expression by Thiostrepton significantly inhibited cell migration/invasion, tumor growth in vitro and in vivo. Collectively, our findings confer that targeting GRB7/ERK/FOXM1 signaling cascade may be a promising molecular therapeutic choice in combating ovarian cancer.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Fatores de Transcrição Forkhead/antagonistas & inibidores , Proteína Adaptadora GRB7/metabolismo , Terapia de Alvo Molecular , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Fosforilação , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA