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











Base de dados
Intervalo de ano de publicação
1.
Bioengineered ; 13(2): 2878-2888, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35040754

RESUMO

Lung adenocarcinoma is the main cause of the excessive mortality for patients who lives with lung cancers. According to the GEPIA database analysis, GATA5 and ARHGAP9 were found to be low expressed in lung adenocarcinoma, and they were positively correlated, and in addition ARHGAP9 low expression was associated with poor prognosis in lung adenocarcinoma. Therefore, the present study focused on the effect of promoting GATA5 to induce ARHGAP9 on the malignant process of lung adenocarcinoma cells. The expressions of GATA5 and ARHGAP9 were measured with Western blot and RT-qPCR. With the adoption of CCK-8, EDU staining, transwell and colony formation, the cell viability, proliferation, invasion and tumorigenesis ability were detected, respectively. In addition, the wound healing and Western blot were employed to evaluate migration and metastasis-related proteins individually. Moreover, the luciferase activity as well as the binding of GATA5 and ARHGAP9 promoters were detected by luciferase report and ChIP. After further comprehensive assessments, the results confirmed that GATA5 could successfully activate ARHGAP9. Moreover, ARHGAP9 upregulation remarkably inhibited lung adenocarcinoma cell proliferation, invasion and migration as compared to the control group. More importantly, GATA5 silencing reversed the inhibitory effect of ARHGAP9 upregulation on the malignant progression of lung adenocarcinoma cells. To conclude, the present study successfully demonstrated for the first time that GATA5-induced ARHGAP9 upregulation has a protective effect on lung adenocarcinoma cells.


Assuntos
Adenocarcinoma de Pulmão/patologia , Fator de Transcrição GATA5/fisiologia , Proteínas Ativadoras de GTPase/genética , Neoplasias Pulmonares/patologia , Células A549 , Adenocarcinoma de Pulmão/diagnóstico , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/mortalidade , Biomarcadores Tumorais/genética , Células Cultivadas , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Prognóstico , Regiões Promotoras Genéticas , Regulação para Cima/genética
2.
Front Immunol ; 12: 704965, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34456915

RESUMO

Interferon-induced transmembrane protein 3 (IFITM3) is an interferon-induced membrane protein, which has been identified as a functional gene in multiple human cancers. The role of IFITM3 in cancer has been preliminarily summarized, but its relationship to antitumor immunity is still unclear. A pancancer analysis was conducted to investigate the expression pattern and immunological role of IFITM3 based on transcriptomic data downloaded from The Cancer Genome Atlas (TCGA) database. Next, correlations between IFITM3 and immunological features in the bladder cancer (BLCA) tumor microenvironment (TME) were assessed. In addition, the role of IFITM3 in estimating the clinical characteristics and the response to various therapies in BLCA was also evaluated. These results were next confirmed in the IMvigor210 cohort and a recruited cohort. In addition, correlations between IFITM3 and emerging immunobiomarkers, such as microbiota and N6-methyladenosine (m6A) genes, were assessed. IFITM3 was enhanced in most tumor tissues in comparison with adjacent tissues. IFITM3 was positively correlated with immunomodulators, tumor-infiltrating immune cells (TIICs), cancer immunity cycles, and inhibitory immune checkpoints. In addition, IFITM3 was associated with an inflamed phenotype and several established molecular subtypes. IFITM3 expression also predicted a notably higher response to chemotherapy, anti-EGFR therapy, and immunotherapy but a low response to anti-ERBB2, anti-ERBB4, and antiangiogenic therapy. In addition, IFITM3 was correlated with immune-related microbiota and m6A genes. In addition to BLCA, IFITM3 is expected to be a marker of high immunogenicity in most human cancers. In conclusion, IFITM3 expression can be used to identify immuno-hot tumors in most cancers, and IFITM3 may be a promising pancancer biomarker to estimate the immunological features of tumors.


Assuntos
Biomarcadores Tumorais/imunologia , Bases de Dados de Ácidos Nucleicos , Regulação Neoplásica da Expressão Gênica/imunologia , Transcriptoma/imunologia , Microambiente Tumoral/imunologia , Neoplasias da Bexiga Urinária/imunologia , Humanos , Inflamação/imunologia , Proteínas de Membrana , Proteínas de Ligação a RNA
3.
Cell Death Dis ; 11(9): 760, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32934206

RESUMO

Palbociclib, a CDK4/6 inhibitor, has been granted accelerated approval by US FDA for hormone receptor-positive HER2-negative metastatic breast cancer. To determine potential biomarkers of palbociclib sensitivity to assist in patient selection and clinical development, we investigated the effects of palbociclib in a panel of molecularly characterized breast cancer cell lines. We quantified palbociclib sensitivity and c-myc expression in 11 breast cancer cell lines, 124 breast cancer samples, and The Cancer Genome Atlas database. We found non-TNBC subtypes were more sensitive to palbociclib than TNBC. Activation of c-myc led to differential palbociclib sensitivities, and further inhibition of c-myc enhanced palbociclib sensitivity. Moreover, we identified for the first time a c-myc/miR-29b-3p/CDK6 axis in breast cancer that could be responsible for c-myc-induced palbociclib insensitivity, in which c-myc activation resulted in downregulation of miR-29b-3p, further activated CDK6 and inhibited cell-cycle arrest at G1 phase. Moreover, downregulated (inactived) c-myc-induced oncogenic addiction could increase palbociclib efficacy, using both Xenograft model and patient-derived tumor xenograft (PDTX) model. Our finding extends the concept of combined blockade of the CDK4/6 and c-myc signaling pathways to increase palbociclib sensitivity, making c-myc a promising biomarker for palbociclib sensitivity in breast cancer.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Quinase 6 Dependente de Ciclina/metabolismo , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Piridinas/farmacologia , Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Quinase 6 Dependente de Ciclina/genética , Feminino , Genes myc , Humanos , Prognóstico , Proteínas Proto-Oncogênicas c-myc/genética
4.
Neoplasia ; 21(6): 533-544, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31029033

RESUMO

Treatment options are limited for patients with triple negative breast cancer (TNBC). Understanding genes that participate in cancer progression and DNA damage response (DDR) may improve therapeutic strategies for TNBC. DAXX, a death domain-associated protein, has been reported to be critically involved in cancer progression and drug sensitivity in multiple cancer types. However, its role in breast cancer, especially for TNBC, remains unclear. Here, we demonstrated a tumor suppressor function of DAXX in TNBC proliferation, colony formation, and migration. In Mouse Xenograft Models, DAXX remarkably inhibited tumorigenicity of TNBC cells. Mechanistically, DAXX could directly bind to the promoter region of RAD51 and impede DNA damage repair, which impacted the protection mechanism of tumor cells that much depended on remaining DDR pathways for cell growth. Furthermore, DAXX-mediated inefficient DNA damage repair could sensitize BRCA-proficient TNBC cells to PARP inhibitors. Additionally, we identified that dual RAD51 and PARP inhibition with RI-1 and ABT888 significantly reduced TNBC growth both in vitro and in vivo, which provided the first evidence of combining RAD51 and PARP inhibition in BRCA-proficient TNBC. In conclusion, our data support DAXX as a modulator of DNA damage repair and suppressor of TNBC progression to sensitize tumors to the PARP inhibitor by repressing RAD51 functions. These provide an effective strategy for a better application of PARP inhibition in the treatment of TNBC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Nucleares/genética , Poli(ADP-Ribose) Polimerase-1/genética , Rad51 Recombinase/genética , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Proteínas Supressoras de Tumor/genética , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Animais , Benzimidazóis/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proteínas Correpressoras , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Feminino , Xenoenxertos , Humanos , Camundongos , Chaperonas Moleculares , Morfolinas/farmacologia , Proteínas Nucleares/antagonistas & inibidores , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Pirróis/farmacologia , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Proteínas Supressoras de Tumor/antagonistas & inibidores
5.
Int J Biol Sci ; 15(3): 522-532, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30745839

RESUMO

The dysregulation of cyclin D -Cyclin-dependent kinase 4/6 (CDK4/6)-Rb axis has been implicated in breast cancer progression and the selective CDK4/6 inhibitors have shown effective activity in advanced breast cancer, especially in tumors driven by the estrogen receptor (ER). However, resistance to these small molecular inhibitors has become an inevitable clinical issue after their initial use. Here, we investigated the potential mechanism of resistance by establishing a CDK4/6 inhibitor palbociclib-resistant breast cancer cell line (MCF-7pR). After prolonged exposure to palbociclib, we detected the loss of the ER signaling and an increase in androgen receptor (AR). Moreover, we demonstrated more localization of AR in the cell nucleus of MCF-7pR compared to the parental cell (MCF-7). We also reported that AR could promote the progression of the cell cycle. Blockade of AR signaling could reduce the level of the relative G1-S cyclins, abolish Rb phosphorylation and inhibit the activation of transcriptional programs in S phase. Furthermore, dual inhibition of AR and CDK4/6 could reverse the resistance of palbociclib both in vitro and in vivo. In sum, our studies provide evidence that AR activation promotes cell cycle progression and cell proliferation in CDK4/6 inhibitor resistance, and identify AR inhibition as a putative novel therapeutic strategy to treat CDK4/6 inhibitor resistance in cancer.


Assuntos
Neoplasias da Mama/metabolismo , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Piperazinas/farmacologia , Piridinas/farmacologia , Receptores Androgênicos/isolamento & purificação , Animais , Benzamidas , Western Blotting , Quinase 4 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/genética , Feminino , Humanos , Imuno-Histoquímica , Imunoprecipitação , Células MCF-7 , Camundongos , Nitrilas , Feniltioidantoína/análogos & derivados , Feniltioidantoína/farmacologia , Receptores Androgênicos/genética
6.
Mol Cancer ; 17(1): 99, 2018 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-30007403

RESUMO

Triple negative breast cancer (TNBC) is a heterogeneous disease with aggressive behavior and poor prognosis. Genomic sequencing has detected a distinctive mutational portrait of both the germline and somatic alterations in TNBC, which is staggeringly different from other breast cancer subtypes. The clinical utility of sequencing germline BRCA1/2 genes has been well established in TNBC. However, for other predisposition genes, studies concerning the risk and penetrance to TNBC are relatively scarce. Very few recurrent mutations, including TP53 and PI3KCA mutations, together with a long tail of individually rare mutations occur in TNBC. These combined effects of genomic alterations drive TNBC progression. Given the complexity and heterogeneity of TNBC, clinical interpretation of the genomic alterations in TNBC may pave a new way for the treatment of TNBC. In this review, we summarized the germline and somatic mutation profiles of TNBC and discussed the current and upcoming therapeutic strategies targeting the mutant proteins or pathways to enable tailored-therapeutics.


Assuntos
Antineoplásicos/uso terapêutico , Análise Mutacional de DNA/métodos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Adulto , Antineoplásicos/farmacologia , Proteína BRCA1/genética , Proteína BRCA2/genética , Ensaios Clínicos como Assunto , Feminino , Mutação em Linhagem Germinativa , Humanos , Pessoa de Meia-Idade , Terapia de Alvo Molecular/métodos , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética
7.
Cell Death Differ ; 25(12): 2209-2220, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29844570

RESUMO

Androgen receptor (AR) is emerging as a novel prognostic biomarker in triple-negative breast cancer (TNBC), but the underlying mechanisms remain unknown. As accumulating evidence has shown that long non-coding RNAs (lncRNAs) regulate important cancer hallmarks, we hypothesised that AR-regulated lncRNAs might play roles in TNBC progression. Here, we performed experiments with or without DHT treatment in three TNBC cell lines, and we identified an AR negatively induced lncRNA (ARNILA), which correlated with poor progression-free survival (PFS) in TNBC patients and promoted epithelial-mesenchymal transition (EMT), invasion and metastasis in vitro and in vivo. Subsequently, we demonstrated that ARNILA functioned as a competing endogenous RNA (ceRNA) for miR-204 to facilitate expression of its target gene Sox4, which is known to induce EMT and contribute to breast cancer progression, thereby promoting EMT, invasion and metastasis of TNBC. Our findings not only provide new insights into the mechanisms of lncRNA in regulating AR but also suggest ARNILA as an alternative therapeutic target to suppress metastasis of TNBC patients.


Assuntos
Neoplasias Mamárias Experimentais/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Receptores Androgênicos/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Animais , Movimento Celular , Feminino , Humanos , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , RNA Longo não Codificante/genética , Neoplasias de Mama Triplo Negativas/diagnóstico por imagem , Células Tumorais Cultivadas
8.
Neoplasia ; 20(5): 478-488, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29605721

RESUMO

Triple negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer that poses a clinical challenge. Thus, new therapy strategies are urgently needed. The selective WEE1 inhibitor, AZD1775, has shown strong anti-proliferative effects on a variety of tumors. Here, we first demonstrate that inhibition of ATR by selective inhibitor AZD6738 can enhance AZD1775-caused growth inhibition in TNBC. Our results show that the enhanced cell death is attributed to repressed DNA damage repair and excessive replication stress, thereby causing increased DNA damage reflected by accumulation of the DNA double-strand-break marker γH2AX. On the other hand, combined treatment with AZD6738 and AZD1775 forces mitotic entry of cells with DNA damages by activating CDK1 activity, inducing severely aberrant mitosis and mitotic catastrophe, ultimately resulting in cell death. Dual inhibition of WEE1 and ATR also inactivated RAD51-mediated homologous recombination, which sensitized TNBC cells to cisplatin and PARP inhibitor. Here, based on the preclinical results that ATR inhibition synergizes with WEE1 inhibition in TNBC, we propose that this combination therapy alone, or in parallel with chemotherapy, represents an innovative and potent targeted therapy in TNBC.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas Nucleares/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Feminino , Humanos , Indóis , Células MCF-7 , Camundongos , Mitose/efeitos dos fármacos , Morfolinas , Pirazóis/farmacologia , Pirimidinas/farmacologia , Pirimidinonas , Sulfonamidas , Sulfóxidos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
9.
Cancer Biol Ther ; 18(6): 369-378, 2017 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-28494179

RESUMO

Treatment of triple negative breast cancer (TNBC) has been a big challenge since it is defined. To date, platinum-based chemotherapy has played a significant role in the treatment of TNBC patients. However, some patients do not respond to platinum salts or gradually develop chemoresistance, resulting in little effect, or even some adverse effects. Here, we review numerous preclinical and clinical investigations to summarize possible mechanisms and potential predictive biomarkers of platinum in TNBC. The homologous recombination deficiency (HRD) resulting from the loss of BRCA function is the main rationale of platinum efficacy in TNBC. BRCA mutation and methylation have been demonstrated to be important potential biomarkers. Based on genome-wide effects, BRCA-like classifier can identify the functional loss of BRCA and work as the predictor. HRD score that is able to identify the "BRCAness" and predict the sensitivity of platinum is increasingly considered. Taken together, all findings suggest that HR deficiency profile encompassed by BRCA mutation and high HRD score could predict response to platinum, even to other DNA-damage inducing agents. p53 family members and molecular subtypes of TNBC are also important alternative considerations for predicting platinum response based on the preclinical trials. Currently, tumor infiltrating lymphocyte level and thrombocytopenia are emerging as predictive biomarkers.


Assuntos
Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/genética , Compostos de Platina/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Antineoplásicos/farmacologia , Reparo do DNA por Junção de Extremidades , Feminino , Instabilidade Genômica , Humanos , Mutação , Compostos de Platina/farmacologia , Resultado do Tratamento , Neoplasias de Mama Triplo Negativas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA