Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Front Med (Lausanne) ; 10: 1167676, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37081842

RESUMO

Background: Breast cancer (BC) is the most common malignant disease worldwide. Although the survival rate is improved in recent years, the prognosis is still bleak once recurrence and metastasis occur. It is vital to investigate more efficient biomarkers for predicting the metastasis and relapse of BC. DYNLT1 has been reported that participating in the progression of multiple cancers. However, there is still a lack of study about the correlation between DYNLT1 and BC. Methods: In this study, we evaluated and validated the expression pattern and prognostic implication of DYNLT1 in BC with multiple public cohorts and BC tumor microarrays (TMAs) of paraffin-embedded tissues collected from the Affiliated Hospital of Jining Medical University. The response biomarkers for immune therapy, such as tumor mutational burden (TMB), between different DYNLT1 expression level BC samples were investigated using data from the TCGA-BRCA cohort utilizing public online tools. In addition, colony formation and transwell assay were conducted to verify the effects of DYNLT1 in BC cell line proliferation and invasion. Results: The results demonstrated that DYNLT1 overexpressed in BC and predicted poor relapse-free survival in our own BC TMA cohort. In addition, DYNLT1 induced BC development by promoting MDA-MB-231 cell proliferation migration, and metastasis. Conclusion: Altogether, our findings proposed that DYNLT1 could be a diagnostic and prognostic indicator in BC.

3.
Cell Res ; 32(4): 359-374, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35197608

RESUMO

TSPAN family of proteins are generally considered to assemble as multimeric complexes on the plasma membrane. Our previous work uncovered that TSPAN8 can translocate into the nucleus as a membrane-free form, a process that requires TSPAN8 palmitoylation and association with cholesterol to promote its extraction from the plasma membrane and subsequent binding with 14-3-3θ and importin-ß. However, what upstream signal(s) regulate(s) the nuclear translocation of TSPAN8, the potential function of TSPAN8 in the nucleus, and the underlying molecular mechanisms all remain unclear. Here, we demonstrate that, epidermal growth factor receptor (EGFR) signaling induces TSPAN8 nuclear translocation by activating the kinase AKT, which in turn directly phosphorylates TSPAN8 at Ser129, an event essential for its binding with 14-3-3θ and importin ß1. In the nucleus, phosphorylated TSPAN8 interacts with STAT3 to enhance its chromatin occupancy and therefore regulates transcription of downstream cancer-promoting genes, such as MYC, BCL2, MMP9, etc. The EGFR-AKT-TSPAN8-STAT3 axis was found to be hyperactivated in multiple human cancers, and associated with aggressive phenotype and dismal prognosis. We further developed a humanized monoclonal antibody hT8Ab4 that specifically recognizes the large extracellular loop of TSPAN8 (TSPAN8-LEL), thus being able to block the extraction of TSPAN8 from the plasma membrane and consequently its nuclear localization. Importantly, both in vitro and in vivo studies demonstrated an antitumor effect of hT8Ab4. Collectively, we discovered an unconventional function of TSPAN8 and dissected the underlying molecular mechanisms, which not only showcase a new layer of biological complexity of traditional membrane proteins, but also shed light on TSPAN8 as a novel therapeutic target for refractory cancers.


Assuntos
Receptores ErbB , Neoplasias , Fator de Transcrição STAT3 , Tetraspaninas , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Humanos , Neoplasias/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Tetraspaninas/genética , Tetraspaninas/metabolismo
4.
Bioengineered ; 12(1): 8965-8979, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34635011

RESUMO

Over the years, molecular subtypes based on estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER-2) status have been observed to effectively guide decision-making for the optimal treatment of patients with breast carcinoma (BRCA). However, despite this progress, there are still more than 41,000 BRCA-related fatalities each year in the United States. Moreover, effective drug targets for triple-negative breast carcinoma (TNBC) are still lacking. Given its high mortality rate, it is necessary to investigate more biomarkers with prognostic and pathological relevance in BRCA. In our study, we examined the expression patterns and prognostic implications of transmembrane P24 trafficking protein 9 (TMED9) in BRCA using multiple public cohorts and BRCA specimens collected from Shanghai General Hospital. In addition to this, in vitro experiments were also performed to evaluate the effects of TMED9 expression in BRCA cell proliferation and migration. Our results have demonstrated that a high expression of TMED9 promoted BRCA cell proliferation and migration and predicted poor prognosis in patients with BRCA. In conclusion, TMED9 is a potential prognostic indicator and a possible drug target of BRCA.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/patologia , Regulação Neoplásica da Expressão Gênica , Proteínas de Transporte Vesicular/metabolismo , Apoptose , Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proliferação de Células , Feminino , Humanos , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas , Proteínas de Transporte Vesicular/genética
5.
Oncogene ; 40(30): 4884-4893, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34163029

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is the deadliest cancer mainly owing to its proclivity to early metastasis and the lack of effective targeted therapeutic drugs. Hence, understanding the molecular mechanisms underlying early invasion and metastasis by PDAC is imperative for improving patient outcomes. The present study identified that upregulation of TSPAN8 expression in PDAC facilitates metastasis in vivo and in vitro. We found SOX9 as a key transcriptional regulator of TSPAN8 expression in response to EGF stimulation. SOX9 modulation was sufficient to positively regulate endogenous expression of TSPAN8, with concomitant in vitro phenotypic changes such as loss of cell-matrix adherence and increased invasion. Moreover, increased SOX9 and TSPAN8 levels were shown to correlate in human pancreatic cancer specimens and downregulated in vitro by EGFR tyrosine kinase inhibitors. High expression of SOX9 and TSPAN8 has been associated with tumor stage, poor prognosis and poor patient survival in PDAC. In conclusion, this study highlights the importance of the EGF-SOX9-TSPAN8 signaling cascade in the control of PDAC invasion and implies that TSPAN8 may be a promising novel therapeutic target for the treatment of PDAC.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias Pancreáticas/etiologia , Neoplasias Pancreáticas/metabolismo , Fatores de Transcrição SOX9/metabolismo , Tetraspaninas/genética , Biomarcadores Tumorais , Movimento Celular/genética , Progressão da Doença , Suscetibilidade a Doenças , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Metástase Neoplásica , Estadiamento de Neoplasias , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Prognóstico , Regiões Promotoras Genéticas , Ligação Proteica , Tetraspaninas/metabolismo
6.
Onco Targets Ther ; 14: 1997-2002, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33776449

RESUMO

INTRODUCTION: Primary tracheal adenoid cystic carcinoma (ACC) is a rare and heterogeneous group of neoplasms arising from the respiratory tract. The solid variant of ACC is a histologically distinct subtype with an unfavorable clinical course. We report on a case of tracheal ACC with immunohistochemical and molecular analysis together with a review of the literature. CASE REPORT: We observed a case in which a 31-year-old male presented with a neoplasm bulging into the lumen and caused symptoms of tracheal obstruction and even hemoptysis. Cytological smears of an endobronchial fine needle aspiration revealed aggregates of basaloid cells with small to medium size, scant cytoplasm, and evenly hyperchromatic nuclei. Histologically, the tumor is characterized by a predominant compact sheet-like and nested pattern of rounded basaloid cells. Immunohistochemically, the tumor was diffusely positive for CK and CD117. CK7 and CK5/6 were focally positive in the genuine glandular structures. P63 was completely negative in the majority of neoplastic cells. Fluorescence in situ hybridization analysis revealed MYB gene rearrangement. CONCLUSION: The solid variant of ACC from trachea is rare and hence poses diagnostic difficulty. Computed tomography (CT) scan and bronchoscopy help assess the extent of the disease. Histological features combined with immunophenotypic and molecular analysis aid in distinguishing this uncommon type from other round cell neoplasms. The accurate diagnosis can help expedite treatment of this highly aggressive tumor.

7.
Aging (Albany NY) ; 11(21): 9778-9793, 2019 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-31727867

RESUMO

Globally, epithelial ovarian cancer (EOC) is the most common gynecological malignancy with poor prognosis. The expression and oncogenic roles of ubiquitin specific peptidase 5 (USP5) have been reported in several cancers except EOC. In the current study, USP5 amplification was highly prevalent in patients with EOC and associated with higher mRNA expression of USP5. USP5 amplification and overexpression was positively correlated with poor prognosis of patients of ovarian serous carcinomas. Disruption of USP5 profoundly repressed cell proliferation by inducing cell cycle G0/G1 phase arrest in ovarian cancer cells. Additionally, USP5 knockdown inhibited xenograft growth in nude mice. Knockdown of USP5 decreased histone deacetylase 2 (HDAC2) expression and increased p27 (an important cell cycle inhibitor) expression in vitro and in vivo. The promoting effects of USP5 overexpression on cell proliferation and cell cycle transition, as well as the inhibitory effects of USP5 overexpression on p27 expression were mediated by HDAC2. Moreover, USP5 interacted with HDAC2, and disruption of USP5 enhanced the ubiquitination of HDAC2. HDAC2 protein was positively correlated USP5 protein, and negatively correlated with p27 protein in ovarian serous carcinomas tissues. Collectively, our data suggest the oncogenic function of USP5 and the potential regulatory mechanisms in ovarian carcinogenesis.


Assuntos
Carcinoma Epitelial do Ovário/enzimologia , Carcinoma Epitelial do Ovário/patologia , Endopeptidases/metabolismo , Histona Desacetilase 2/metabolismo , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia , Idoso , Animais , Carcinógenos/metabolismo , Carcinoma Epitelial do Ovário/genética , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Endopeptidases/genética , Feminino , Amplificação de Genes , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Neoplasias Ovarianas/genética , Prognóstico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ubiquitinação
8.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 47(5): 499-506, 2018 05 25.
Artigo em Chinês | MEDLINE | ID: mdl-30693692

RESUMO

OBJECTIVE: To investigate the anti-inflammatory effect and mechanisms of interleukin-35 (IL-35) in inflammatory bowel disease. METHODS: BALB/c mice were divided into three groups with 10 mice in each group:control group, model group (oral administration of 4% glucan sodium sulfate for 7 d) and IL-35-treated group (oral administration of 4% glucan sodium sulfate for 7 d, intraperitoneal injection of 2 µg IL-35 at d2-5). Disease activity index (DAI) was scored every day. After 7 d, the mice were sacrificed, and the serum and intestinal tissue samples were collected. The gross morphology of the colon was observed; HE staining was used to observe the pathological changes of colon tissue; flow cytometry was employed to detect the change of macrophage polarization ratio in colon tissue; the mRNA expression levels of cytokines IL-6, TNF-α, IFN-γ, IL-10 and SHIP1 in colon tissue were determined by real-time quantitative RT-PCR; the expression and distribution of SHIP1 in colon tissue was measured by immunohistochemistry; Western blotting was adopted to detect the expression level of SHIP1 protein in colonic intestinal tissues of each group. RESULTS: The DAI scores of the mice in the model group were higher than those in the control group, while the DAI scores in the IL-35-treated group were lower than those in the model group (all P<0.01). Compared with the control group, the colon length was significantly shortened in the model group (P<0.05), while the colon length of the IL-35-treated group had an increasing trend compared with the model group, but the difference was not statistically significant (P>0.05). Compared with the model group, microscopic inflammatory infiltration score was decreased and microscopic crypt destruction and score was significantly lower in IL-35-treated group (all P<0.05). The relative expression of proinflammatory cytokines IL-6, TNF-α and IFN-γ in the colon tissue of IL-35-treated group was decreased compared with the model group, while the relative expression of IL-10 mRNA was higher than that of the model group (all P<0.05). Compared with the control group, the proportion of M1 macrophages in the model group increased (P<0.05), while the proportion of M1 macrophages in the IL-35-treated group was lower than that in the model group (P<0.05). The relative expression of SHIP1 mRNA and protein in the colon tissue of IL-35-treated group was higher than that in the model group (all P<0.05). CONCLUSIONS: IL-35 can inhibit the polarization of M1 macrophages and regulate inflammatory cytokines to promote anti-inflammatory effect on mice with colitis.


Assuntos
Anti-Inflamatórios , Colite , Colo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Interleucinas , Animais , Anti-Inflamatórios/farmacologia , Colite/tratamento farmacológico , Colite/fisiopatologia , Colo/efeitos dos fármacos , Citocinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Glucanos/farmacologia , Interleucina-6/genética , Interleucinas/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases/genética
9.
Mol Cancer Ther ; 15(8): 1943-1951, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27196763

RESUMO

The tumor microenvironment impacts tumor progression and individual cells, including CD4(+) T cells, which have been detected in bladder cancer tissues. The detailed mechanism of how these T cells were recruited to the bladder cancer tumor and their impact on bladder cancer progression, however, remains unclear. Using a human clinical bladder cancer sample survey and in vitro coculture system, we found that bladder cancer has a greater capacity to recruit T cells than surrounding normal bladder tissues. The consequences of higher levels of recruited T cells in bladder cancer included increased bladder cancer metastasis. Mechanism dissection revealed that infiltrating T cells might function through secreting the cytokine IL1, which increases the recruitment of T cells to bladder cancer and enhances the bladder cancer androgen receptor (AR) signaling that results in increased bladder cancer cell invasion via upregulation of hypoxia-inducible factor-1α (HIF1α)/VEGFa expression. Interruption of the IL1→AR→HIF1α→VEGFa signals with inhibitors of HIF1α or VEGFa partially reversed the enhanced bladder cancer cell invasion. Finally, in vivo mouse models of xenografted bladder cancer T24 cells with CD4(+) T cells confirmed in vitro coculture studies and concluded that infiltrating CD4(+) T cells can promote bladder cancer metastasis via modulation of the IL1→AR→HIF1α→VEGFa signaling. Future clinical trials using small molecules to target this newly identified signaling pathway may facilitate the development of new therapeutic approaches to better suppress bladder cancer metastasis. Mol Cancer Ther; 15(8); 1943-51. ©2016 AACR.


Assuntos
Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Neoplasias da Bexiga Urinária/imunologia , Neoplasias da Bexiga Urinária/metabolismo , Animais , Linhagem Celular Tumoral , Quimiotaxia de Leucócito/imunologia , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Interleucina-1/metabolismo , Masculino , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Neoplasias da Bexiga Urinária/patologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Oncol Rep ; 35(6): 3409-18, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27109614

RESUMO

Non-small cell lung cancer (NSCLC) as the most frequently diagnosed lethal cancer remains the major cause of overall cancer-related death worldwide. Testes-specific protease 50 (TSP50) has been proved as a critical biomarker in various cancers, and we previously reported that TSP50 protein expression is overexpressed in clinical resected NSCLC tumor tissues and related to poor prognosis in NSCLC patients. Hence, the present study was designed to further investigate the potential oncogenesis mechanism of TSP50 in NSCLC cells. Real-time quantitative PCR, immunohistochemical assay and western blot analysis were used to analyze the TSP50 mRNA and protein expression in 20 NSCLC cases, and TSP50 expression was observed to have high levels in the NSCLC specimens and paired metastatic lymph node tissues when compared to the levels in corresponding normal lung tissues and normal lymph nodes. In the experiments in NSCLC cell lines, lentiviral short hairpin RNA (shRNA) delivery system was applied to knock down TSP50 in 95D cells, and the following investigations revealed that downregulation of TSP50 expression markedly reduced cell proliferation, colony formation and migration ability in vitro. Furthermore, the inhibition of TSP50 induced G0/G1-phase arrest and decreased expression levels of cell cycle relative markers CDK4, CDK6, and CyclinD1 and increased expression of p21 and p53 in 95D cells. In conclusion, this study indicates that TSP50 plays a significant role in NSCLC cell proliferation and may act as a novel oncogene in the development and progression of NSCLC, offering a potential cancer therapeutic target for the treatment of NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Pontos de Checagem do Ciclo Celular , Neoplasias Pulmonares/patologia , Serina Endopeptidases/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Lentivirus/genética , Metástase Linfática , RNA Interferente Pequeno/genética , Serina Endopeptidases/genética , Proteína Supressora de Tumor p53/fisiologia
11.
Biochem Biophys Res Commun ; 464(1): 147-53, 2015 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-26102039

RESUMO

Ubiquitin-like with PHD and RING finger domain 1 (UHRF1) is a multi-domain ubiquitin E3 ligase that plays critical roles in regulation of DNA methylation and histone ubiquitination. In this study, we found UHRF1 is frequently overexpressed in human clear cell Renal Cell Carcinoma (ccRCC) tissues both at mRNA and protein levels. We showed that UHRF1 directly interacts with p53 both in vivo and in vitro. A new domain (PD) in UHRF1 was required for interaction with p53. We found that UHRF1 down-regulates p53 transactivation activity which was depends on the ubiquitin E3 ligase function. UHRF1 can promote non-degradative ubiquitination of p53, suppress p53 pathway activation and p53-dependent apoptosis in ccRCC cells. Together, our study suggests that UHRF1, which overexpressed ccRCC, may act as a p53 regulator, suppress p53 pathway activation and help ccRCC cells to escape from p53-dependent apoptosis.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , Carcinoma de Células Renais/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Renais/genética , Proteína Supressora de Tumor p53/genética , Apoptose , Proteínas Estimuladoras de Ligação a CCAAT/antagonistas & inibidores , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Dano ao DNA , Células HEK293 , Humanos , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Ligação Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Ativação Transcricional , Transfecção , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA