Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Genet ; 15: 1296570, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38510272

RESUMO

Background: Ulcerative colitis (UC) is a common and progressive inflammatory bowel disease primarily affecting the colon and rectum. Prolonged inflammation can lead to colitis-associated colorectal cancer (CAC). While the exact cause of UC remains unknown, this study aims to investigate the role of the TWIST1 gene in UC. Methods: Second-generation sequencing data from adult UC patients were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified, and characteristic genes were selected using machine learning and Lasso regression. The Receiver Operating Characteristic (ROC) curve assessed TWIST1's potential as a diagnostic factor (AUC score). Enriched pathways were analyzed, including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Variation Analysis (GSVA). Functional mechanisms of marker genes were predicted, considering immune cell infiltration and the competing endogenous RNA (ceRNA) network. Results: We found 530 DEGs, with 341 upregulated and 189 downregulated genes. TWIST1 emerged as one of four potential UC biomarkers via machine learning. TWIST1 expression significantly differed in two datasets, GSE193677 and GSE83687, suggesting its diagnostic potential (AUC = 0.717 in GSE193677, AUC = 0.897 in GSE83687). Enrichment analysis indicated DEGs associated with TWIST1 were involved in processes like leukocyte migration, humoral immune response, and cell chemotaxis. Immune cell infiltration analysis revealed higher rates of M0 macrophages and resting NK cells in the high TWIST1 expression group, while TWIST1 expression correlated positively with M2 macrophages and resting NK cell infiltration. We constructed a ceRNA regulatory network involving 1 mRNA, 7 miRNAs, and 32 long non-coding RNAs (lncRNAs) to explore TWIST1's regulatory mechanism. Conclusion: TWIST1 plays a significant role in UC and has potential as a diagnostic marker. This study sheds light on UC's molecular mechanisms and underscores TWIST1's importance in its progression. Further research is needed to validate these findings in diverse populations and investigate TWIST1 as a therapeutic target in UC.

2.
Support Care Cancer ; 32(2): 97, 2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38200158

RESUMO

PURPOSE: Malignant ascites (MA) often occurs in recurrent abdominal malignant tumors, and the large amount of ascites associated with cancerous peritonitis not only leads to severe abdominal distension and breathing difficulties, but also reduces the patient's quality of life and ability to resist diseases, which usually makes it difficult to carry out anti-cancer treatment. The exploration of MA treatment methods is also a key link in MA treatment. This article is going to review the treatment of MA, to provide details for further research on the treatment of MA, and to provide some guidance for the clinical treatment of MA. METHOD: This review analyzes various expert papers and summarizes them to obtain the paper. RESULT: There are various treatment methods for MA, including systemic therapy and local therapy. Among them, systemic therapy includes diuretic therapy, chemotherapy, immunotherapy, targeted therapy, anti angiogenic therapy, CAR-T, and vaccine. Local therapy includes puncture surgery, peritoneal vein shunt surgery, acellular ascites infusion therapy, radioactive nuclide intraperitoneal injection therapy, tunnel catheter, and intraperitoneal hyperthermia chemotherapy. And traditional Chinese medicine treatment has also played a role in enhancing efficacy and reducing toxicity to a certain extent. CONCLUSION: Although there has been significant progress in the treatment of MA, it is still one of the clinical difficulties. Exploring the combination or method of drugs with the best therapeutic effect and the least adverse reactions to control MA is still an urgent problem to be solved.


Assuntos
Carcinoma , Neoplasias Peritoneais , Humanos , Ascite/etiologia , Ascite/terapia , Qualidade de Vida , Recidiva Local de Neoplasia , Imunoterapia , China
3.
Cell Commun Signal ; 20(1): 96, 2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-36058938

RESUMO

BACKGROUND: The cyclin-dependent kinase 7 (CDK7) inhibitor THZ1 represses multiple cancer cells. However, its tumor-repressive efficiency in wild-type p53 breast cancer cells remains controversial. METHODS: We conducted various assays, including CCK8, colony formation, flow cytometry, western blotting, and lactate dehydrogenase release detection, to clarify whether p53 elevation sensitizes breast cancer cells to THZ1. RESULTS: We found that upregulating functional p53 contributes to the increased sensitivity of breast cancer cells to THZ1. Increased THZ1 sensitivity requires active p53 and an intact p53 pathway, which was confirmed by introducing exogenous wild-type p53 and the subsequent elevation of THZ1-mediated tumor suppression in breast cancer cells carrying mutant p53. We confirmed that p53 accumulates in the nucleus and mitochondria during cell death. Furthermore, we identified extensive transcriptional disruption, rather than solely CDK7 inhibition, as the mechanism underlying the nutlin-3 and THZ1-induced death of breast cancer cells. Finally, we observed the combined nutlin-3 and THZ1 treatment amplified gasdermin E cleavage. CONCLUSION: Enhanced sensitivity of breast cancer cells to THZ1 can be achieved by increasing effective p53 expression. Our approach may serve as a potential treatment for patients with breast cancer resistant to regular therapies. Video Abstract.


Assuntos
Neoplasias da Mama , Proteína Supressora de Tumor p53 , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Fenilenodiaminas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Proteína Supressora de Tumor p53/metabolismo
4.
J Oncol ; 2022: 5901450, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35794989

RESUMO

Objective: To investigate the clinical efficacy and safety of albumin paclitaxel combined with intrapleural bevacizumab + lobaplatin for patients with non-squamous non-small cell lung cancer (NS-NSCLC) with malignant pleural effusion (MPE) and analyze prognostic factors. Methods: A total of 126 NS-NSCLC patients were included in the study. Control group with 64 cases received intrapleural infusion of lobaplatin + intravenous albumin paclitaxel, and treatment group with 62 cases received additional intrapleural bevacizumab perfusion. Analysis was performed by collecting data about MPE, progression-free survival (PFS), overall survival (OS), and scores of quality of life. Results: In the treatment and control groups, objective response rate (ORR) was 51.6% and 31.3% (χ 2 = 5.39, P=0.02), and disease control rate (DCR) was 91.9% and 71.9% (χ 2 = 8.49, P=0.004), respectively. The main adverse reactions (≥grade 3) in the treatment group were thrombocytopenia, peripheral neurotoxicity, proteinuria, neutropenia, and nausea/vomiting, and in the control group, they were weakness, nausea/vomiting, anemia, and peripheral neurotoxicity. In the control and treatment groups, the median PFS was 6.2 (95% confidence interval (CI): 5.86-6.56) and 5.1 (95% CI: 4.956-5.191), and the median OS was 14.4 (95% CI: 12.681-16.113) and 10.6 months (95% CI: 8.759-12.391). The score of quality of life for treated patients was significantly higher than those before treatment and the control group, and the parameters included general health status (GH), role physical (RP), body pain (BP), social function (SF), and vitality (VT); pH, CD4+/CD8+ values, and vascular endothelial growth factor (VEGF) in the pleural effusion significantly affected the PFS and OS (P < 0.05). Bevacizumab administration in patients with bloody pleural effusion did not increase the risk of pleural hemorrhage. Conclusion: The combination of albumin paclitaxel and intrapleural bevacizumab + lobaplatin is effective and may reverse the adverse events in patients with NS-NSCLC and MPE. The change of CD4+/CD8+ ratio before and after treatment is an independent and prognostic factor for patients with NS-NSCLC and MPE.

5.
Eur J Med Res ; 27(1): 121, 2022 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-35842733

RESUMO

BACKGROUND: Colorectal cancer (CRC) is one of the leading causes of cancer-related death worldwide. P21 activated kinase 4 (PAK4) and Breast cancer anti-estrogen resistance 3 (BCAR3) have been reported to be involved in numerous aspects in tumorous progression. In this study, we propose to screen multi-targeted microRNAs. (miRNAs), which simultaneously inhibit neoplastic evolution through suppressing the transcription of target genes. METHODS: MTT and Colony formation assays measured cell's viability and proliferation. Scratch wound and Transwell assays detected the ability in migration and invasion for SW116 cells. The multi-targeted microRNAs of PAK4 and BCAR3 were predicted using bioinformatics analysis and verified by conducting dual luciferase reporter assay, western blot and qRT-PCR that could detect the expression levels of miR-199a/b-3p. RESULTS: The knockdown of PAK4 significantly impeded proliferation and colony formation of SW1116 cells when the knockdown of BCAR3 hindered migration and invasion of SW1116 cells. MiR-199a/b-3p directly targeted the 3'-UTR of PAK4 and BCAR3, further effected proliferation, colony formation, migration, and invasion of SW1116 cells. PAK4 or BCAR3 overexpression could partially reversed inhibitory effects of miR-199a/b-3p. CONCLUSIONS: These results provided a new multi-targeted cite for cancerous suppressant to improve the prognosis of CRC inpatients.


Assuntos
Neoplasias Colorretais , MicroRNAs , Regiões 3' não Traduzidas/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fatores de Troca do Nucleotídeo Guanina/farmacologia , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo , Quinases Ativadas por p21/farmacologia
6.
Front Mol Biosci ; 8: 697457, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34490348

RESUMO

Higher cyclin-dependent kinase (CDK7) expression is a character of breast cancer and indicates poor prognosis. Inhibiting CDK7 exhibited effective cancer cell suppression which implies the potential of CDK7 inhibition to be a method for anti-cancer treatment. Our study aimed to explore a novel mechanism of CDK7 inhibition for suppressing breast cancer cell survival. Here, we proved inhibiting CDK7 repressed breast cancer cell proliferation and colony formation and increased the apoptotic cell rate, with p53 and GSDME protein level elevation. When p53 was suppressed in MCF-7 cells, the decline of GSDME expression and associated stronger proliferation and colony formation could be observed. Since downregulation of GSDME was of benefit to breast cancer cells, p53 inhibition blocked the elevation of GSDME induced by CDK7 inhibition and retrieved cells from the tumor suppressive effect of CDK7 inhibition. Therefore, CDK7 inhibition exerted a negative effect on breast cancer cell proliferation and colony formation in a p53-GSDME dependent manner. These results revealed the CDK7-p53-GSDME axis could be a pathway affecting breast cancer cell survival.

7.
Ann Palliat Med ; 10(5): 5754-5762, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33977736

RESUMO

BACKGROUND: To explore the clinical efficacy, safety, and prevention of major adverse reactions of the non-steroidal anti-inflammatory drug celecoxib combined with OxyContin and Pregabalin in the treatment of cancerous pudendal neuralgia. METHODS: A total of 51 patients presenting with pelvic malignancies with cancerous pudendal neuralgia were selected, and random number table method was used to allocate them to either the experimental group (n=27) or control group (n=24). The control group was treated with OxyContin combined with Pregabalin, and the experimental group was treated with Celecoxib on the basis of the control group. RESULTS: At 24 hours after treatment, the clinical effective rate of the experimental group was 92.6%, which was significantly higher than the 66.7% of the control group (P<0.05). The numerical rating scale (NRS) scores of the 2 groups of participants on the 7th and 14th days after treatment were lower than before treatment (P<0.05), and the NRS scores of the participants in the experimental group had decreased more significantly. At the same time, the average daily consumption of OxyContin on the 7th and 14th day of the experimental group was lower than that of the control group (P<0.05). Compared with the control group, the incidence of constipation and dysuria in the experimental group was significantly reduced (P<0.05). Co-occurring in both groups during treatment, 10 participants with urinary dysfunction were treated with tamsulosin hydrochloride sustained-release capsules, no urinary retention occurred, catheterization was avoided, tamsulosin hydrochloride sustained-release capsules could be stopped after 1 week, and urination was smooth (P<0.05). After treatment, the quality of life of the 2 groups of participants had improved compared to before treatment, and the improvement was more significant in the experimental group. CONCLUSIONS: When treating patients with cancerous pudendal neuralgia with OxyContin and Pregabalin, the addition of celecoxib has a significant effect, which can effectively improve the patient's pain, improve their quality of life to a certain extent, and reduce the consumption of OxyContin. Lowering the dose of OxyContin reduces the occurrence of adverse reactions related to the drug, especially the incidence of constipation and urinary retention. Tamsulosin hydrochloride sustained-release capsules can effectively relieve urinary disorders caused by OxyContin. TRIAL REGISTRATION: Chinese Clinical Trial Registry ChiCTR2100046045.


Assuntos
Preparações Farmacêuticas , Neuralgia do Pudendo , Humanos , Pregabalina/efeitos adversos , Qualidade de Vida , Resultado do Tratamento
8.
Biophys Rep ; 7(2): 142-151, 2021 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-37288149

RESUMO

Human umbilical cord mesenchymal stem cells (HUC-MSCs) are pluripotent and functional in many biological processes, by which releasing secretary factors to promote the self-repairing of damaged tissue or developing into functional cell at local organ. However, there is a high risk that oxidative stress would reduce the pluripotency and factor-secretion during the preparation and transplantation. Therefore, reducing oxidative stress is expected to improve the efficacy of HUC-MSCs therapy. Zinc (Zn) is an essential trace element which involves in the resistance of oxidative stress. To investigate Zn-regulated signaling pathways, we have profiled the gene expression at transcriptome level in primary HUC-MSCs treated with zinc sulfate, followed with GO and KEGG gene enrichment analysis. Zn treatment improved signal pathways for mineral absorption, cell growth, and cell death. Zn deficiency was mimicked by TPEN administration, which suppressed cell proliferation and reduced the expression of HUC-MSCs surface stem cell markers CD73, CD90 and CD105 by flow cytometry. Nuclear factor erythrocyte 2 related factor 2 (Nrf2) plays an important role in antioxidant biological processes. In vitro treatment of Zn significantly increased Nrf2 and Sirt3 expression at gene level and protein level respectively. Zn supplementation inhibited TPEN-induced failure of cell survival and reversed the reduction of Nrf2 and Sirt3 expression, which further reduced the production of ROS. Zn successfully presented its anti-oxidation effect by activating Nrf2/Sirt3 signaling pathway in HUC-MSCs. Zn supplementation may improve the efficacy of HUC-MSCs therapy with reduced oxidative stress.

9.
Arch Biochem Biophys ; 689: 108415, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32562663

RESUMO

Regorafenib, a multiple kinase inhibitor, is recently approved for treatment of patients with advanced hepatocellular carcinoma (HCC). Previous studies demonstrated that regorafenib was a mitochondrial toxicant, which associated with the impairment of mitochondria. Sirt3 is involved in the regulation of mitochondrial function in cancers. This study aimed to investigate the mechanism of Sirt3 involved in the mitochondrial dysfunction which associated with regorafenib treatment in liver cancer cells. We found regorafenib inhibited Sirt3 and p-ERK expression in HCC cells in a dose-dependent manner. Bioinformatics analysis showed that Sirt3 expression was down-regulated in liver cancer tissues and its low expression was correlated with worse overall survival (OS) in liver cancer patients. After transfected with Sirt3 overexpression plasmid, we found that Sirt3 sensitized liver cancer cells to regorafenib and resulted in much more apoptosis with a significant increase of ROS level. However, exogenous antioxidant could not weaken the apoptosis. Mitochondrial membrane potential assay indicated that Sirt3 overexpression accelerated the mitochondrial depolarization process induced by regorafenib and aggravated mitochondrial injury. Cellular oxygen consumption assay showed that mitochondrial dysfunction was caused by the damage of the electron transport chain. The results demonstrated that Sirt3 overexpression promoted the increase of ROS and apoptosis induced by regorafenib through the acceleration of mitochondrial dysfunction by impairing function of the electron transport chain in liver cancer cells. Our studies verified the functional role of Sirt3 in regorafenib treatment and suggested that regorafenib accompanied with Sirt3 activator as a novel treatment strategy for HCC.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Sirtuína 3/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Humanos , Neoplasias Hepáticas/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo
10.
Am J Physiol Cell Physiol ; 318(3): C640-C648, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31940246

RESUMO

Zinc (Zn) has antioxidant effect in different types of organs and is closely associated with human health. Endometrial receptivity is one of the most important factors in the embryo implantation and development. However, the regulatory mechanism of Zn in endometrium tissue is still unclear. In the study, we found that plasma Zn level is significantly associated with female infertility, which severely affects female reproductive health. Primary endometrial stromal cells were isolated from female endometrium and cultured in the laboratory. Zn chelator TPEN treatment reduced the expression of stem cell markers CD73, CD90, and CD105 and generated reactive oxygen species in endometrial stromal cells. However, pretreatment of Zn (zinc sulfate) is able to prevent TPEN-induced oxidative stress in vitro. By transcriptional profiling and gene ontology analysis, we found that Zn increased the cellular pluripotency signaling and extracellular matrix-receptor interaction, but reduced autophagy, endocytosis, and the nitrogen metabolism pathway. We further discovered the antioxidant function of Zn through the peroxisome proliferator-activated receptor gamma coactivator 1α/nuclear factor erythroid-2-related factor signaling pathway in endometrial stromal cells. Zn supplementation may open up an effective therapeutic approach for patients with oxidative stress-related endometrial diseases.


Assuntos
Endométrio/metabolismo , Fator 2 Relacionado a NF-E2/fisiologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/fisiologia , Células Estromais/metabolismo , Transcrição Gênica/fisiologia , Zinco/metabolismo , Adulto , Sobrevivência Celular/fisiologia , Células Cultivadas , Endométrio/diagnóstico por imagem , Feminino , Humanos , Transdução de Sinais/fisiologia , Células Estromais/patologia , Adulto Jovem
11.
Biomed Pharmacother ; 104: 729-737, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29807222

RESUMO

Cripto-1 is an oncogenic protein that belongs to the epidermal growth factor (EGF)-cripto-1/FRL1/cryptic (CFC) family. It has been shown to stimulate tumorigenesis and metastasis by promoting cancer cell proliferation, epithelial-to-mesenchymal transition (EMT), and tumor angiogenesis. However, the role of Cripto-1 in cell survival and apoptosis remains largely undefined. In the present study, we found that Cripto-1 is significantly upregulated in a number of human cancer cell lines. The membrane-associated but not the soluble form of Cripto-1 promotes resistance to drug-induced caspase-3 cleavage, an indicator of apoptosis. Consequently, Cripto-1 silencing sensitizes human cancer cells to chemotherapy drugs including cytarabine, cisplatin and taxol. Our mechanistic studies revealed that Cripto-1 promotes apoptosis resistance by inducing NF-κB-mediated Survivin expression through activation of TAK-1. We also found that Cripto-1 silencing does not affect growth of un-treated cancer cells, and Cripto-1 forms self-assembled punctiforms and changes its subcellular distribution upon cytarabine treatment. Thus, the anti-apoptotic activity of Cripto-1 could be an inducible function that can be activated by external stimuli such as drug stimulation. Our findings suggested that targeting the Cripto-1/TAK-1/NF-κB/Survivin pathway may be an effective approach to combat apoptosis resistance in cancer.


Assuntos
Apoptose/fisiologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Proteínas Ligadas por GPI/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , MAP Quinase Quinase Quinases/genética , NF-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Transdução de Sinais/fisiologia , Apoptose/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , Carcinogênese/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/farmacologia , Citarabina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Fator de Crescimento Epidérmico/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Células HEK293 , Humanos , Neovascularização Patológica/metabolismo , Paclitaxel/farmacologia , Transdução de Sinais/efeitos dos fármacos , Survivina
12.
Int J Biochem Cell Biol ; 73: 63-71, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26855338

RESUMO

Previous studies have shown that testes-specific protease 50 (TSP50), a pro-oncogene overexpressed in many types of tumors, could promote cell proliferation, invasion, tumorigenesis, and tumor metastasis, suggesting that it is a potential cancer therapeutic target in drug discovery. Here, a luciferase assay system driven by the TSP50 gene promoter was used to screen the inhibitor of expression of TSP50. The study found that cardamonin, a flavone compound, could efficiently inhibit the expression of TSP50 in both mRNA and protein levels. Further results revealed that cardamonin also efficiently inhibited the viability of TSP50 high-expressing cancer cells by inducing G2/M-phase arrest and mitochondrial-dependent apoptosis. Surprisingly, knocking down the expression of TSP50 gene had the same effects as treatment with cardamonin. Moreover, it has been found that cardamonin had an inhibitory potency on TSP50 high-expressing tumor growth in vivo. In contrast, overexpression of TSP50 greatly decreased the cell sensitivity to the inhibitory effect of cardamonin and reversed the decreased tumor-inhibitory effect of cardamonin. Additionally, both TSP50 interference and treatment with cardamonin could suppress p65 nuclear translocation, and overexpression of TSP50 reversed the suppressive effect of cardamonin on p65 nuclear translocation. Taken together, these results suggest that cardamonin inhibited cell viability and tumorigenesis at least partially via blocking the activation of TSP50-mediated nuclear factor-kappaB signaling pathway, and cardamonin may be a promising anticancer drug candidate in the development of a novel agent for TSP50 high-expressing cancer cells.


Assuntos
Chalconas/farmacologia , NF-kappa B/metabolismo , Testículo/enzimologia , Apoptose/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Células HEK293 , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Regiões Promotoras Genéticas/genética , Serina Endopeptidases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Testículo/efeitos dos fármacos
13.
Tumour Biol ; 37(3): 3405-15, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26449823

RESUMO

Hepatitis B virus (HBV) is implicated in the pathogenesis of hepatocellular carcinoma, which has been found to be associated with TGF-beta signaling. Activin A is a TGF-ß family cytokine that exhibits cell proliferation inhibition on normal hepatocyte. How HBV-encoded X oncoprotein play in activin's activity on hepatocyte has not been developed. In this study, a nontumor hepatic cell line HL7702 with HBX ectogenic expression has been established. MTT and BrdU assays showed that HBx promoted growth of HL7702 cells in vitro and downregulated activin signaling. Deregulated activin signaling pathway by HBX failed to activate target gene p21/waf1 and p15 transcription. In addition, mammalian two-hybrid and coimmunoprecipitation assays revealed that HBX could directly interact with activin signaling transduction protein Smad4, making activated Smad2/3/4 nucleus translocation suppressed. Furthermore, we detected that leptomycin B, the inhibitor of CRM1 protein, could recover nuclear translocation of endogenous Smads complex in HL7702 with HBX expression, indicating that HBX antagonized Smads nucleus translocation, at least partially, on CRM1-dependent manner. Leptomycin B was found to have antigrowth activity on HBX-expressed HL7702, according to its antitumor function in previous study. Above all, HBX antagonized activin signaling in normal human liver cells by interacting with Smad4 might one of the considerable causes of HBX-induced hepatocyte transformation, which deprived activin's cell growth inhibition function at an early stage of tumorigenesis.


Assuntos
Ativinas/fisiologia , Hepatócitos/fisiologia , Carioferinas/fisiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Proteína Smad4/fisiologia , Transativadores/fisiologia , Transporte Ativo do Núcleo Celular , Proliferação de Células , Células Cultivadas , Humanos , Neoplasias Hepáticas/etiologia , Proteínas Virais Reguladoras e Acessórias , Proteína Exportina 1
14.
IUBMB Life ; 67(10): 768-77, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26399456

RESUMO

MicroRNA-199a/b-3p is downregulated in several types of aggressive cancer, and its decrement significantly correlates with poor survival. Here, we aim to investigate the biological function of miR-199a/b-3p and its regulation of target genes in breast cancer cells with highly metastatic potential. In addition, we found that miR-199a/b-3p expression was much lower in MDA-MB-231, CAL120, and HCC1395 breast cancer cells with highly metastatic potential. Functional assays showed that restored miR-199a/b-3p expression inhibited MDA-MB-231 cell growth, cell-cycle progression, migration, and invasion. In addition, we experimentally demonstrated that PAK4 was the direct target of miR-199a/b-3p, hypo-expression of PAK4 suppressed proliferation, migration and invasion of MDA-MB-231 cells, and overexpression of PAK4 significantly rescued the inhibitory effect of miR-199a/b-3p on MDA-MB-231 cell growth, migration, and invasion. Further, we also observed that miR-199a/b-3p could inactivate the PAK4/MEK/ERK signaling pathway. Thus, miR-199a/b-3p functions as a tumor suppressor and has an important role in breast cancer metastasis through PAK4/MEK/ERK signaling pathway.


Assuntos
Sistema de Sinalização das MAP Quinases , MicroRNAs/fisiologia , Quinases Ativadas por p21/metabolismo , Regiões 3' não Traduzidas , Sequência de Bases , Sítios de Ligação , Neoplasias da Mama , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação para Baixo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Invasividade Neoplásica , Interferência de RNA , Quinases Ativadas por p21/genética
15.
Toxicol Lett ; 224(3): 349-55, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24252419

RESUMO

Testes-specific protease 50 (TSP50) is aberrantly expressed in many cancer biopsies and plays a crucial role in tumorigenesis, which make it a potential cancer therapeutic target for drug discovery. Here, we constructed a firefly luciferase reporter driven by the TSP50 gene promoter to screen natural compounds capable of inhibiting the expression of TSP50. Then we identified alantolactone, a sesquiterpene lactone, could efficiently inhibit the promoter activity of TSP50 gene, further results revealed that alantolactone also efficiently inhibited the expression of TSP50 in both mRNA and protein levels. Moreover, we found alantolactone could increase the ratio of Bax/Bcl-2, and activate caspase-9 and caspase-3 in the cancer cells with high expression of TSP50, surprisingly, the same effects can also be observed in the same cells just by knockdown of TSP50 gene expression. Furthermore, our results suggested that overexpression of TSP50 decreased the cell sensitivity to alantolactone-induced apoptosis in those cancer cells. Taken together, these results suggest that alantolactone induces mitochondrial-dependent apoptosis at least partially via down-regulation of TSP50 expression.


Assuntos
Antifúngicos/toxicidade , Apoptose/efeitos dos fármacos , Lactonas/toxicidade , Serina Endopeptidases/biossíntese , Serina Endopeptidases/efeitos dos fármacos , Sesquiterpenos de Eudesmano/toxicidade , Western Blotting , Caspase 3/biossíntese , Caspase 9/biossíntese , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/ultraestrutura , Regulação para Baixo/efeitos dos fármacos , Humanos , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Plasmídeos/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Serina Endopeptidases/genética , Transfecção , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
16.
Biochem J ; 436(2): 457-67, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21385156

RESUMO

TSP50 (testes-specific protease 50) is a testis-specific expression protein, which is expressed abnormally at high levels in breast cancer tissues. This makes it an attractive molecular marker and a potential target for diagnosis and therapy; however, the biological function of TSP50 is still unclear. In the present study, we show that overexpression of TSP50 in CHO (Chinese-hamster ovary) cells markedly increased cell proliferation and colony formation. Mechanistic studies have revealed that TSP50 can enhance the level of TNFα (tumour necrosis factor α)- and PMA-induced NF-κB (nuclear factor κB)-responsive reporter activity, IκB (inhibitor of NF-κB) α degradation and p65 nuclear translocation. In addition, the knockdown of endogenous TSP50 in MDA-MB-231 cells greatly inhibited NF-κB activity. Co-immunoprecipitation studies demonstrated an interaction of TSP50 with the NF-κB-IκBα complex, but not with the IKK (IκB kinase) α/ß-IKKγ complex, which suggested that TSP50, as a novel type of protease, promoted the degradation of IκBα proteins by binding to the NF-κB-IκBα complex. Our results also revealed that TSP50 can enhance the expression of NF-κB target genes involved in cell proliferation. Furthermore, overexpression of a dominant-negative IκB mutant that is resistant to proteasome-mediated degradation significantly reversed TSP50-induced cell proliferation, colony formation and tumour formation in nude mice. Taken together, the results of the present study suggest that TSP50 promotes cell proliferation, at least partially, through activation of the NF-κB signalling pathway.


Assuntos
Proliferação de Células , NF-kappa B/metabolismo , Serina Endopeptidases/fisiologia , Transdução de Sinais/fisiologia , Animais , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , NF-kappa B/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Chem Commun (Camb) ; 47(10): 2940-2, 2011 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-21258749

RESUMO

Folate-functionalized polyoxometalate nanoparticles have unique oxidase-like activity, which can facilitate the fast oxidation of organic dyes without using any oxidizing agents or peroxidases especially at neutral pH conditions. This nanoparticle could be used as an agent in colorimetric multiplexed immunoassays.


Assuntos
Materiais Biomiméticos/química , Colorimetria/métodos , Ácido Fólico/química , Imunoensaio/métodos , Molibdênio/química , Oxirredutases/metabolismo , Vanádio/química , Benzidinas/química , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Modelos Moleculares , Conformação Molecular , Nanopartículas/química , Oxirredução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA