Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Mol Carcinog ; 63(6): 1092-1105, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38415870

RESUMO

Colorectal cancer (CRC) is a common digestive tract tumor with the third incidence and death in the world. There is still an urgent need for effective therapeutic targets and prognostic markers for CRC. Herein, we report a novel potential target and marker, Chordin like-1 (CHRDL1). The function of CHRDL1 has been reported in gastric cancer, breast cancer, and oral squamous cell carcinoma. However, the biological effect of CHRDL1 in CRC remains unrevealed. Transwell and tube formation experiments were used to determine the biological function of CHRDL1. Western blot and rescue experiments were used to determine the specific mechanisms of CHRDL1. Results showed CHRDL1 is significantly downregulated in CRC cell lines and tissues. In vitro, experiments confirmed that CHRDL1 can inhibit cell growth, migration, invasion, angiogenesis and reverse epithelial-mesenchymal transformation. In vivo, experiments proved that it can inhibit tumor growth and metastasis. Mechanistically, we newly find that CHRDL1 exerts biological functions through the transforming growth factor-beta (TGF-ß)/vascular endothelial growth factor signaling axis in vitro and in vivo. Therefore, we concluded that CHRDL1 reduces the growth, migration, and angiogenesis of CRC cells by downregulating TGF-ß signaling. Our new findings on CHRDL1 may provide a basis for clinical antiangiogenesis therapy and the prognosis of CRC.


Assuntos
Movimento Celular , Proliferação de Células , Neoplasias Colorretais , Transição Epitelial-Mesenquimal , Neovascularização Patológica , Transdução de Sinais , Fator de Crescimento Transformador beta , Fator A de Crescimento do Endotélio Vascular , Humanos , Neoplasias Colorretais/patologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/genética , Animais , Neovascularização Patológica/patologia , Neovascularização Patológica/metabolismo , Camundongos , Fator de Crescimento Transformador beta/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Camundongos Nus , Feminino , Metástase Neoplásica , Ensaios Antitumorais Modelo de Xenoenxerto , Camundongos Endogâmicos BALB C , Angiogênese
2.
Biochem Biophys Res Commun ; 664: 108-116, 2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-37141638

RESUMO

Pancreatic cancer is an extremely aggressive malignancy with a very disappointing prognosis. Neuroglobin (NGB), a member of the globin family, has been demonstrated to have a significant role in a variety of tumor forms. The possible role of NGB as a tumor suppressor gene in pancreatic cancer was investigated in this work. Information from the public dataset TCGA combined with GTEx was used to analyze the finding that NGB was commonly downregulated in pancreatic cancer cell lines and tissues, correlating with patient age and prognosis. The expression of NGB in pancreatic cancer was investigated via RT-PCR, qRT-PCR, and Western blot experiments. In-vitro and in-vivo assays, NGB elicited cell cycle arrest in the S phase and apoptosis, hindered migration and invasion, reversed the EMT process, and suppressed cell proliferation and development. The mechanism of action of NGB was predicted via bioinformatics analysis and validated using Western blot and co-IP experiments revealed that NGB inhibited the EGFR/AKT/ERK pathway by binding to and reducing expression of GNAI1 and p-EGFR. In addition, pancreatic cancer cells overexpressing NGB showed increased drug sensitivity to gefitinib (EGFR-TKI). In conclusion, NGB inhibits pancreatic cancer progression by specifically targeting the GNAI1/EGFR/AKT/ERK signaling axis.


Assuntos
Neuroglobina , Neoplasias Pancreáticas , Proteínas Proto-Oncogênicas c-akt , Humanos , Linhagem Celular Tumoral , Proliferação de Células , Receptores ErbB/genética , Receptores ErbB/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroglobina/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
3.
Dokl Biochem Biophys ; 499(1): 273-281, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34426926

RESUMO

BACKGROUND: Chemotherapy drugs commonly used for cancer therapy, but chemotherapy has limitation due to side effects. Current studies suggest natural products are reducing the side effects of chemotherapy medicines. In this study, we examined the side effects of doxorubicin (Dox) in esophageal cancer cells (CSCs) derived tumors in vivo. METHODS: Esophageal cancer cells (YMI) were treated in vitro with daphnetin (DAP) along with DOX. The MTT assay was used for estimating the cell viability and Annexin/7-AAD was used for the determination of apoptosis. Cell cycle arrest was conducted using the PI-staining method. The potential effect of DAP was evaluated by the estimation of oxidative stress such as total antioxidant capacity (TAC), malondialdehyde (MDA) and superoxide dismutase (SOD) and body weight in the xenograft mice. RESULTS: DAP can protect Dox cell toxicity by suppressing cell apoptosis of ESCC. DAP arrest the cells as S-phase. In vivo experimental study showed that Dox simultaneously with DAP decreases the tumor size along with increased body weight in the nude mice compared to Dox alone treated group mice. Dox along with the DAP exhibited less systemic toxicity and reduced oxidative stress fraction circulation. CONCLUSION: The result suggests that daphnetin may be used as an adjuvant therapy to reduce the systemic toxicity of chemotherapeutic agents, such as DOX, in stem cell treatment with ESCC cancer.


Assuntos
Antineoplásicos/efeitos adversos , Doxorrubicina/efeitos adversos , Neoplasias Esofágicas/patologia , Células-Tronco Neoplásicas/patologia , Umbeliferonas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Masculino , Malondialdeído/metabolismo , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos
4.
Dig Liver Dis ; 56(1): 176-186, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37230858

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) is a highly vascularized tumor with a poor prognosis. Novel vascular-related therapeutic targets and prognostic markers remain urgently needed. AIMS: To investigate the role and mechanism of CLCA1 in hepatocellular carcinoma. METHODS: Immunofluorescence, Co-immunoprecipitation and rescue experiment were used to determine the specific mechanisms of CLCA1. Chemosensitivity assay was used to measure the impact of CLCA1 on Sorafenib. RESULTS: CLCA1 was dramatically downregulated in hepatocellular carcinoma cell lines and tissues. Ectopic expression of CLCA1 induced cell apoptosis and G0/G1 phase arrest while suppressed cell growth, inhibited migration and invasion, reversal of epithelial mesenchymal transition in vitro and reduced xenograft tumor growth in vivo. Mechanistically, CLCA1 could co-localize and interact with TGFB1, thereby suppressing HCC angiogenesis through the TGFB1/SMAD/VEGF signaling cascade in vitro and in vivo. Moreover, CLCA1 also enhanced the sensitivity of HCC cells to the first-line targeted therapy, Sorafenib. CONCLUSION: CLCA1 sensitizes HCC cells to Sorafenib and suppresses hepatocellular carcinoma angiogenesis through downregulating TGFB1 signaling cascade. This newly identified CLCA1 signaling pathway may help guide the anti-angiogenesis therapies for hepatocellular carcinoma. We also support the possibility of CLCA1 being a prognostic biomarker for hepatocellular carcinoma.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Sorafenibe/farmacologia , Sorafenibe/uso terapêutico , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Angiogênese , Linhagem Celular Tumoral , Proliferação de Células , Apoptose , Fator de Crescimento Transformador beta1/metabolismo , Canais de Cloreto/uso terapêutico
5.
Dig Liver Dis ; 2023 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-37968146

RESUMO

BACKGROUND: Colorectal cancer (CRC) is one of the most commonly diagnosed malignant tumours of the digestive tract, and new therapeutic targets and prognostic markers are still urgently required. However, the role and molecular mechanisms of ATP binding cassette subfamily A member 8 (ABCA8) in CRC remain unclear. METHODS: Databases and clinical specimens were analysed to determine the expression level of ABCA8 in CRC. Colony formation, CCK-8 and Transwell assays were conducted to determine cell proliferation, viability, migration and invasion. Flow cytometry was used to detect cell cycle progression and apoptosis. Western blot and rescue experiments were performed to determine the specific mechanisms of action of ABCA8. RESULTS: ABCA8 expression is dramatically down-regulated in CRC tissues and cell lines. Ectopic expression of ABCA8 induced apoptosis and cell cycle arrest in vitro, inhibited cell growth, suppressed migration and invasion, reversed epithelial-mesenchymal transition and suppressed xenograft tumour growth and metastasis in vivo. Mechanistically, ABCA8 inhibited CRC cell proliferation and metastasis through the Wnt/ß-catenin signalling pathway, both in vitro and in vivo. CONCLUSION: We verified that ABCA8 inhibits the malignant progression of CRC through the Wnt/ß-catenin pathway. This newly discovered ABCA8-Wnt-ß-catenin signalling axis is probably helpful in guiding the clinical diagnosis and treatment of CRC.

6.
Adv Clin Exp Med ; 30(9): 957-966, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34358419

RESUMO

BACKGROUND: Gastric cancer (GC) is a common malignant tumor with a high morbidity and mortality worldwide. It has been reported that V-Myb avian myeloblastosis viral oncogene homolog-like 2 (MYBL2) could be a promising prognostic biomarker for GC. However, the specific role of MYBL2 in GC progression remains unclear. OBJECTIVES: To examine the role of MYBL2 in GC progression and investigate the underlying mechanisms. MATERIAL AND METHODS: The mRNA levels of target genes were detected using quantitative real-time polymerase chain reaction (RT-qPCR) and protein expression was measured with western blot analysis. Cell Counting Kit-8 (CCK-8) and colony formation assays were employed to inspect HGC-27 cell proliferation, and cellular apoptosis was determined with TUNEL staining. Finally, the interaction of MYBL2 and cell division cycle 20 (CDC20) was verified with immunoprecipitation. RESULTS: MYBL2 was confirmed to be overexpressed in GC cells. MYBL2 knockdown inhibited HGC-27 cell proliferation and promoted cellular apoptosis, and these effects were reversed by CDC20 overexpression. Interestingly, MYBL2 interacted with CDC20 and regulated its expression. MYBL2 knockdown also inhibited activation of the Wnt/ß-catenin signaling pathway, while CDC20 overexpression showed the opposite effect. CONCLUSIONS: In summary, the synergy between MYBL2 and CDC20 induced the proliferation of GC cells and inhibited cell apoptosis; these effects may have involved the Wnt/ß-catenin signaling pathway. Thus, MYBL2 may be a promising target for GC treatment.


Assuntos
Proteínas Cdc20 , Proteínas de Ciclo Celular , Neoplasias Gástricas , Transativadores , Apoptose , Proteínas Cdc20/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Neoplasias Gástricas/genética , Transativadores/genética , Via de Sinalização Wnt
7.
Asian Pac J Cancer Prev ; 15(11): 4689-95, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24969905

RESUMO

BACKGROUND: A great number of studies have shown that cytochrome P450 1A1 (CYP1A1) genetic polymorphisms, CYP1A1 Msp I and CYP1A1 Ile/Val, might be risk factors for digestive tract cancers, including esophageal cancer (EC), gastric cancer (GC), hepatic carcinoma (HC), as well as colorectal cancer (CC), but the results are controversial. In this study, a meta-analysis of this literature aimed to clarify associations of CYP1A1 genetic polymorphisms with digestive tract cancer susceptibility in Chinese populations. MATERIALS AND METHODS: Eligible case-control studies published until December 2013 were retrieved by systematic literature searches from PubMed, Embase, CBM, CNKI and other Chinese databases by two investigators independently. The associated literature was acquired through deliberate search and selection based on established inclusion criteria. Fixed- effects or random-effects models were used to estimate odds ratios (ORs and 95%CIs). The meta-analysis was conducted using Review Manager 5.2 and Stata 12.0 softwares with stability evaluated by both stratified and sensitivity analyses. Moreover, sensitivity analysis and publication bias diagnostics confirmed the reliability and stability. RESULTS: Eighteen case control studies with 1, 747 cases and 2, 923 controls were selected for CYP1A1 MspI polymorphisms, and twenty case-control studies with 3, 790 cases and 4, 907 controls for the CYP1A1 Ile/ Val polymorphisms. Correlation associations between CYP1A1 Ile/Val polymorphisms and digestive tract cancers susceptibility were observed in four genetic models in the meta-analysis (GG vs AA:OR= 2.03, 95%CI =1.52- 2.72; AG vs AA: OR=1.26, 95%CI =1.07-1.48; [ GG+AG vs AA] :OR =1.42, 95%CI=1.20-1.68, [GG vs AA+AG ]:OR=1.80, 95%CI =1.40-2.31). There was no association between CYP1A1 Msp I polymorphisms and digestive tract cancer risk. Subgroup analysis for tumor type showed a significant association of CYP1A1 Ile/Val genetic polymorphisms with EC in China. However, available data collected by the study failed to reveal remarkable associations of GC or HC with CYP1A1 Ile/Val genetic polymorphisms and EC, GC or CC with CYP1A1 MspI genetic polymorphisms. CONCLUSIONS: Our results indicated that CYP1A1 Ile/Val genetic polymorphisms, but not CYP1A1 Msp I polymorphisms, are associated with an increased digestive tract cancer risk in Chinese population. Additional well-designed studies, with larger sample size, focusing on different ethnicities and cancer types are now warranted to validate this finding.


Assuntos
Povo Asiático/genética , Citocromo P-450 CYP1A1/genética , Neoplasias do Sistema Digestório/etiologia , Neoplasias do Sistema Digestório/genética , Predisposição Genética para Doença/genética , Polimorfismo Genético/genética , Estudos de Casos e Controles , Genótipo , Humanos , Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA