Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
J Exp Clin Cancer Res ; 42(1): 274, 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37864196

RESUMO

BACKGROUND: Tumor cells can resist chemotherapy-induced pyroptosis through glycolytic reprogramming. Estrogen-related receptor alpha (ERRα) is a central regulator of cellular energy metabolism associated with poor cancer prognosis. Herein, we refine the oncogenic role of ERRα in the pyroptosis pathway and glycolytic metabolism. METHODS: The interaction between ERRα and HIF-1α was verified using co-immunoprecipitation. The transcriptional binding sites of ERRα and NLRP3 were confirmed using dual-luciferase reporter assay and cleavage under targets and tagmentation (CUT&Tag). Flow cytometry, transmission electron microscopy, scanning electron microscopy, cell mito stress test, and extracellular acidification rate analysis were performed to investigate the effects of ERRα on the pyroptosis pathway and glycolytic metabolism. The results of these experiments were further confirmed in endometrial cancer (EC)-derived organoids and nude mice. In addition, the expression of ERRα-related pyroptosis genes was analyzed using The Cancer Genome Atlas and Gene Expression Omnibus database. RESULTS: Triggered by a hypoxic microenvironment, highly expressed ERRα could bind to the promoter of NLRP3 and inhibit caspase-1/GSDMD signaling, which reduced inflammasome activation and increased pyroptosis resistance, thereby resulting in the resistance of cancer cells to cisplatin. Moreover, ERRα activated glycolytic rate-limiting enzyme to bridge glycolytic metabolism and pyroptosis in EC. This phenomenon was further confirmed in EC-derived organoids and nude mice. CUT & Tag sequencing and The Cancer Genome Atlas database analysis showed that ERRα participated in glycolysis and programmed cell death, which resulted in EC progression. CONCLUSIONS: ERRα inhibits pyroptosis in an NLRP3-dependent manner and induces glycolytic metabolism, resulting in cisplatin resistance in EC cells.


Assuntos
Neoplasias do Endométrio , Proteína 3 que Contém Domínio de Pirina da Família NLR , Humanos , Camundongos , Animais , Feminino , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Caspase 1/genética , Caspase 1/metabolismo , Caspase 1/farmacologia , Camundongos Nus , Piroptose , Cisplatino/farmacologia , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Glicólise , Microambiente Tumoral , Proteínas de Ligação a Fosfato/genética , Proteínas de Ligação a Fosfato/metabolismo , Proteínas de Ligação a Fosfato/farmacologia , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptor ERRalfa Relacionado ao Estrogênio
2.
J Transl Med ; 21(1): 204, 2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36932403

RESUMO

BACKGROUND: Endometrial cancer (EC) is one of the most common gynecological malignancies globally, and the development of innovative, effective drugs against EC remains a key issue. Phytoestrogen kaempferol exhibits anti-cancer effects, but the action mechanisms are still unclear. METHOD: MTT assays, colony-forming assays, flow cytometry, scratch healing, and transwell assays were used to evaluate the proliferation, apoptosis, cell cycle, migration, and invasion of both ER-subtype EC cells. Xenograft experiments were used to assess the effects of kaempferol inhibition on tumor growth. Next-generation RNA sequencing was used to compare the gene expression levels in vehicle-treated versus kaempferol-treated Ishikawa and HEC-1-A cells. A network pharmacology and molecular docking technique were applied to identify the anti-cancer mechanism of kaempferol, including the building of target-pathway network. GO analysis and KEGG pathway enrichment analysis were used to identify cancer-related targets. Finally, the study validated the mRNA and protein expression using real-time quantitative PCR, western blotting, and immunohistochemical analysis. RESULTS: Kaempferol was found to suppress the proliferation, promote apoptosis, and limit the tumor-forming, scratch healing, invasion, and migration capacities of EC cells. Kaempferol inhibited tumor growth and promotes apoptosis in a human endometrial cancer xenograft mouse model. No significant toxicity of kaempferol was found in human monocytes and normal cell lines at non-cytotoxic concentrations. No adverse effects or significant changes in body weight or organ coefficients were observed in 3-7 weeks' kaempferol-treated animals. The RNA sequencing, network pharmacology, and molecular docking approaches identified the overall survival-related differentially expressed gene HSD17B1. Interestingly, kaempferol upregulated HSD17B1 expression and sensitivity in ER-negative EC cells. Kaempferol differentially regulated PPARG expression in EC cells of different ER subtypes, independent of its effect on ESR1. HSD17B1 and HSD17B1-associated genes, such as ESR1, ESRRA, PPARG, AKT1, and AKR1C1\2\3, were involved in several estrogen metabolism pathways, such as steroid binding, 17-beta-hydroxysteroid dehydrogenase (NADP+) activity, steroid hormone biosynthesis, and regulation of hormone levels. The molecular basis of the effects of kaempferol treatment was evaluated. CONCLUSIONS: Kaempferol is a novel therapeutic candidate for EC via HSD17B1-related estrogen metabolism pathways. These results provide new insights into the efficiency of the medical translation of phytoestrogens.


Assuntos
Neoplasias do Endométrio , Estradiol Desidrogenases , Quempferóis , Farmacologia em Rede , Animais , Feminino , Humanos , Camundongos , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Estrogênios/metabolismo , Quempferóis/farmacologia , Simulação de Acoplamento Molecular , PPAR gama/metabolismo , Esteroides/metabolismo , Estradiol Desidrogenases/metabolismo
3.
J Inflamm Res ; 15: 6153-6163, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36386586

RESUMO

Purpose: This study aimed to explore lymphocyte subsets for the personalized prediction of endometrioid endometrial cancer (EEC) risk and evaluated the correlation between immune cells and International Federation of Gynecology and Obstetrics (FIGO) staging in patients with EEC. Patients and Methods: A case-control study was conducted to analyze the clinical data of 421 patients admitted to Fujian Maternity and Child Health Hospital from October 2017 to December 2021. t-tests or Mann-Whitney U-tests were used to analyze the percentages and absolute counts of peripheral blood lymphocyte subsets in patients with EEC and patients without cancer. The independent risk factors for ECC and FIGO stage were analyzed via multivariate binary logistic regression. The receiver operating characteristic curve was calculated to evaluate the prediction efficacy of risk factors on ECC. Results: The CD4+ T% in the 121 patients with EEC was lower than in the 300 patients without cancer (P = 0.013). The absolute counts of peripheral CD4+ T (P = 0.002) and T cells (P = 0.007) in 37 patients with EEC were lower than in 51 patients without cancer. Multivariate binary logistic regression analysis showed that CD4+ T% and natural killer cell (NK)% were independent risk factors for FIGO staging in patients with EEC. NK% was significantly higher in patients with advanced stage (FIGO III and IV) than those with early EEC (FIGO I and II) (P = 0.004). To determine the early and advance FIGO stage of EEC, the cutoff value of NK% was calculated as 19.94%. Conclusion: With the decrease of CD4+ T counts, the immune status of patients with EEC is impaired. NK cells may help in the evaluation of the prognosis of patients with EEC and are likely to be an independent risk factor for FIGO staging in patients with EEC.

4.
Front Oncol ; 12: 881252, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35800058

RESUMO

Oxygen is critical to energy metabolism, and tumors are often characterized by a hypoxic microenvironment. Owing to the high metabolic energy demand of malignant tumor cells, their survival is promoted by metabolic reprogramming in the hypoxic microenvironment, which can confer tumor cell resistance to pyroptosis. Pyroptosis resistance can inhibit anti-tumor immunity and promote the development of malignant tumors. Hypoxia inducible factor-1α (HIF-1α) is a key regulator of metabolic reprogramming in tumor cells, and estrogen-related receptor α (ERRα) plays a key role in regulating cellular energy metabolism. Therefore, the close interaction between HIF-1α and ERRα influences the metabolic and functional changes in cancer cells. In this review, we summarize the reprogramming of tumor metabolism involving HIF-1α/ERRα. We review our understanding of the role of HIF-1α/ERRα in promoting tumor growth adaptation and pyroptosis resistance, emphasize its key role in energy homeostasis, and explore the regulation of HIF-1α/ERRα in preventing and/or treating endometrial carcinoma patients. This review provides a new perspective for the study of the molecular mechanisms of metabolic changes in tumor progression.

5.
Tumour Biol ; 34(3): 1667-78, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23430583

RESUMO

Bladder cancer (BC) is one of the most common human malignancies that account for major death in the world. Apoptin that is derived from chicken anemia virus (CAV) has displayed tumor-specific cytotoxic activity in a variety of human carcinomas. However, the magical function of apoptin in bladder carcinoma cell lines has not been identified yet. In our study, we delivered apoptin into bladder-originating T24, EJ, and HCV29 cell lines by adenovirus system. The selective cytotoxic effect of apoptin was determined by cell viability assay, active caspase-3 measurement, and annexin V/PI double staining. Importantly, we have examined the differential expression patterns of tumor-associated genes including Ki67, C-erbB-2, Rb, and nm23 by flow cytometry and western blot in vitro. In an animal study, apoptin was infused into animal models by AAV system, and immunohistochemistry and quantitative real-time PCR (qRT-PCR) were employed to validate results in vivo. The results indicated that apoptin could selectively induce apoptosis in bladder tumorigenic cells coupled with tumor-specific nucleus accumulation in vitro. Interestingly, apoptin could downregulate expression levels of Ki67 and C-erbB-2 and upregulate the expression of Rb both in vitro and in vivo. Moreover, the animal models treated with AAV-apoptin have shown smaller tumor volumes and displayed better prognosis than controls. In conclusion, apoptin could selectively induce apoptosis in bladder tumor cells through altering expression profiles of tumor-associated genes.


Assuntos
Apoptose , Biomarcadores Tumorais/metabolismo , Proteínas do Capsídeo/metabolismo , Proliferação de Células , Transcriptoma , Neoplasias da Bexiga Urinária/patologia , Animais , Biomarcadores Tumorais/genética , Western Blotting , Proteínas do Capsídeo/genética , Vírus da Anemia da Galinha , Dependovirus/genética , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Genes erbB-2/genética , Vetores Genéticos , Humanos , Técnicas Imunoenzimáticas , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nucleosídeo NM23 Difosfato Quinases/genética , Nucleosídeo NM23 Difosfato Quinases/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida , Transplante Homólogo , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/mortalidade , Neoplasias da Bexiga Urinária/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA