Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Gene Med ; 26(5): e3686, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38689382

RESUMO

BACKGROUND: The cell endocrine pathway is a critical physiological process composed of the endoplasmic reticulum, Golgi apparatus and associated vesicles. Loss of enzymes or proteins can cause dysfunction of endoplasmic reticulum and Golgi apparatus and affect secretion pathways leading to a variety of human diseases, including cancer. METHODS: The single-cell RNA sequencing and single nucleotide variant principal component analysis data of ovarian cancer were retrieved from The Cancer Genome Atlas and Gene Expression Omnibus (GEO) datasets. Eighty-four genes from SECRETORY_PATHWAYs were obtained from the gene set enrichment analysis (GSEA) website. Univariate cox regression analyses and ConsensusClusterPlus were used to identify prognostic genes and molecular subtypes, which were validated using the tumor immune dysfunction and exclusion (i.e. TIDE) analysis and gene mutation analysis. A prognosis model was established by randomForestSRC. Abundant infiltrated immune cells and pathway enrichment analyses were carried out, respectively, through ssGSEA, ESTIMATE, MCP-counter and GSEA. The drug sensitive analysis was performed using pRRophetic package. Immunotherapy datasets and pan-carcinoma analysis were used to examine the performance of prognostic model. RESULTS: Eighteen prognostic genes from SECRETORY_PATHWAYs were found in both TCGA and GEO datasets. Next, two clusters (C1 and C2) were determined, for which C1 with a poor prognosis had higher immune infiltration. Tumor-related pathways, such as PATHWAYS_IN_CANCER and B_CELL_RECEPTOR_SIGNALING_PATHWAY, were enriched in C1. Moreover, C2 was suitable for immunotherapy. A four-gene (DNAJA1, NDRG3, LUZP1 and ZCCHC24) signature was developed and successfully validated. RiskScore of higher levels were significantly associated with worse prognoses. An enhanced immune infiltration, increased pathways score and inappropriate immunotherapy were observed in the high RiskScore group. The high- and low-RiskScore groups had different drug sensitivities. Immunotherapy datasets and pan-carcinoma analysis indicated that the low RiskScore group may benefit from immunotherapy. CONCLUSIONS: Based on the perspective of the secretory signaling pathway, a robust prognostic signature with great performances was determined, which may provide clues for clinical precision treatment of ovarian cancer.


Assuntos
Biomarcadores Tumorais , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Neoplasias Ovarianas , Humanos , Feminino , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Prognóstico , Biomarcadores Tumorais/genética , Transcriptoma , Biologia Computacional/métodos , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
2.
Front Cell Dev Biol ; 9: 699487, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34307378

RESUMO

In the past decade, several discoveries have documented the existence of innervation in ovarian cancer and cervical cancer. Notably, various neurotransmitters released by the activation of the sympathetic nervous system can promote the proliferation and metastasis of tumor cells and regulate immune cells in the tumor microenvironment. Therefore, a better understanding of the mechanisms involving neurotransmitters in the occurrence and development of gynecological cancers will be beneficial for exploring the feasibility of using inexpensive ß-blockers and dopamine agonists in the clinical treatment of gynecological cancers. Additionally, this article provides some new insights into targeting tumor innervation and neurotransmitters in the tumor microenvironment.

3.
Oncol Rep ; 46(1)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34080660

RESUMO

Cervical cancer is considered one of the diseases with the highest mortality among women and with limited treatment options. Hydrogen (H2) inhalation has been reported to have a variety of tumor­suppressive effects, but the exact mechanism remains unclear. In the present study, HeLa cervical cancer cells and HaCaT keratinocytes treated with H2, and a HeLa xenograft mouse model subjected to H2 inhalation were established. TUNEL, Cell Counting Kit­8 and Ki67 staining assays were used to detect cell apoptosis and proliferation. Oxidative stress was determined according to the levels of reactive oxygen species, malondialdehyde and superoxide dismutase. Tumor growth was recorded every 3 days, and the excised tumors were stained with hematoxylin and eosin. High­throughput RNA sequencing and subsequent Gene Ontology (GO) enrichment analysis were performed in HeLa­treated and un­treated HeLa cells. The expression of hypoxia­inducible factor (HIF)­1α and NF­κB p65 was verified by western blotting, immunohistochemistry and reverse transcription­quantitative PCR. The results revealed an increased apoptosis rate, and reduced cell proliferation and oxidative stress in H2­treated HeLa cells but not in HaCaT cells. Similarly, decreased tumor growth and cell proliferation, and enhanced cell apoptosis were observed in H2­treated HeLa tumors. RNA sequencing and GO analysis suggest that downregulated HIF1A (HIF­1α mRNA) and RelA (NF­κB p65) levels, and reduced NF­κB signaling were associated with the antitumor effect of H2. Finally, decreased HIF­1α and NF­κB p65 expression both at the transcriptional and translational levels were observed in H2­treated HeLa cells and in HeLa­derived tumors. In conclusion, the present study reveals a novel mechanism of H2 against cervical cancer, which may serve as a potential therapeutic target in clinical practice.


Assuntos
Antineoplásicos/administração & dosagem , Perfilação da Expressão Gênica/métodos , Hidrogênio/administração & dosagem , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Fator de Transcrição RelA/genética , Neoplasias do Colo do Útero/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Hidrogênio/farmacologia , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Análise de Sequência de RNA , Resultado do Tratamento , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cancer Manag Res ; 13: 3485-3492, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33911898

RESUMO

PURPOSE: To compare perioperative outcomes between robotic single-site surgical technique and conventional laparoendoscopic single-site surgical technique. METHODS: This was a retrospective cohort study involving 67 patients who received robotic single-site surgery or laparoendoscopic single-site surgery for the treatment of stage IB1 cervical squamous carcinoma. The robotic single-site radical hysterectomy technique combined with pelvic lymph node dissections were performed in 32 patients while the laparoendoscopic single-site radical hysterectomy technique combined with pelvic lymph node dissections were performed in 35 patients. RESULTS: The enrolled patients had been diagnosed with stage IB1 cervical squamous carcinoma. The perioperative outcomes were mean age (51.63±8.32 years in the lymph node dissection (RSS group) and 53.14±8.14 years in the lymph node dissection (LESS group), p=0.453); BMIs (23.76±2.72 in the RSS group and 23.46±2.28 in the LESS group, p=0.629); shorter operative times (223.56±15.43 min in the RSS group and 248.61±20.89 min in the LESS group, p<0.01) and less estimated blood loss (217.25±16.77 mL in the RSS group and 294.74±24.00 mL in the LESS group, p<0.01). None of the study participants exhibited postoperative pain. There were no statistically significant differences in the length of hospital stay (p=0.865), perioperative complications (p=0.602), duration of closure and removal of catheter (p=0.518) as well as in pathological diagnoses between the two groups. CONCLUSION: Robotic single-site surgery can be used in the treatment of early stage cervical cancer as it exhibits acceptable operative times and perioperative outcomes. This surgical technique is feasible and safe.

5.
Cell Biol Int ; 45(5): 1030-1037, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33404163

RESUMO

Antimetastatic effect of Metformin has been documented in epithelial ovarian cancer (EOC). Presently, we investigated the regulatory mechanism of Metformin in EOC metastasis. First, Girdin was significantly enhanced in EOC tumorous tissues and cell lines. Seconded, knockdown of Girdin significantly suppressed EOC cell viability, migration, and invasion, while upregulation of Girdin produced the opposite effects in vitro and facilitated lung metastasis in EOC cell xenograft in vivo. In addition, we confirmed that the inhibitory effect of Metformin on Girdin expression. Mechanistically, the oncogenic effects of Girdin could be reversed by LY294002 (an AKT pathway inhibitor) and Metformin. These results suggested that Metformin attenuated EOC metastasis through Girdin and targeting Girdin may be a promising therapeutic strategy for EOC in the future.


Assuntos
Carcinoma Epitelial do Ovário/metabolismo , Proteínas dos Microfilamentos/genética , Metástase Neoplásica/tratamento farmacológico , Proteínas de Transporte Vesicular/genética , Adulto , Animais , Carcinoma Epitelial do Ovário/genética , Carcinoma Epitelial do Ovário/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Sobrevivência Celular/genética , Transição Epitelial-Mesenquimal , Feminino , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Metformina/metabolismo , Metformina/farmacologia , Camundongos Nus , Proteínas dos Microfilamentos/efeitos dos fármacos , Proteínas dos Microfilamentos/metabolismo , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Transcriptoma/genética , Proteínas de Transporte Vesicular/efeitos dos fármacos , Proteínas de Transporte Vesicular/metabolismo
6.
Int J Clin Exp Pathol ; 11(8): 4230-4235, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-31949818

RESUMO

OBJECTIVE: Specific sgRNA-sequences targeting oncogenes E6 and E7 in HPV18 were designed using the CRISPR/Cas9 system. These sgRNAs knocked out E6 and E7 expressions and were used to study their effects on the proliferation and cell cycle of the cervical cancer HeLa cell line. METHODS: Lentivirus vectors targeting E6 and E7 oncogenes were constructed and transfected into HeLa cells. mRNA and protein expression levels of E6 and E7 were measured by RT-PCR and Western blot, respectively. The cell cycle was detected by flow cytometry. A colony formation assay was applied to evaluate the proliferation capacity of the HeLa cells. RESULTS: Three E6 Cas9-sgRNA vectors targeting E6 and three E7 Cas9-sgRNA vectors targeting E7 genes were constructed and transfected into HeLa cells, respectively. RT-PCR results showed that all three E6 and E7 sgRNAs inhibited the expressions of E6 or E7 mRNA, respectively, when compared with the control groups. The inhibition ratios of the three groups of E6-sgRNAs were 28%, 85%, and 19%; the E7-sgRNAs were 86%, 25%, and 27%, respectively (P<0.05), with E6-sgRNA2 and E7-sgRNA1 having the greatest inhibitory effects. Western blot results showed that, compared with the control group, the protein expressions of E6 and E7 in the sgRNAs transfected group were also decreased, and E6-sgRNA2 and E7-sgRNA1 had the most inhibitory effects on E6 and E7 proteins. Flow cytometry results showed that the number of cells in G1/G0 was increased by 14.2% in the E6-sgRNA2 transfection group, and by 7.1% in the E7-sgRNA1 transfection group. Colony formation assay results showed that after transfection of E6 or E7 sgRNA plasmids, the HeLa cell colony was reduced significantly compared with the control group. CONCLUSIONS: The CRISPR/Cas9 system targeting HPV18 E6 or E7 genes effectively blocked the transcription and expression of oncogenes E6 or E7 in HeLa cells, which resulted in cell cycle arrest and reduced cell proliferation.

7.
Oncol Lett ; 14(6): 7557-7564, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29344202

RESUMO

Metformin protects against insulin resistance by restoring insulin sensitivity and may also possess anticancer activity. The aim of the present study was to investigate the effects of metformin alone or combined with cisplatin (DDP) on the cell viability and apoptosis of HO-8910 human ovarian cancer cells, and to investigate metformin as a potential novel therapeutic for treating ovarian cancer. The viability of HO-8910 cells was assessed using a cell proliferation and cytotoxicity assay following treatment with different concentrations of metformin (0.01, 0.5, 1, 5 and 10 mM) and/or 5 µM of DDP. Flow cytometry was performed to examine cell apoptosis, and western blotting was used to measure the expression of extracellular signal-related kinase 1/2 (ERK1/2) phosphorylated (p)-ERK1/2, vascular endothelial growth factor (VEGF), VEGF receptor 2 (VEGFR2), B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X (Bax) and caspase-3. The resultsof the present study revealed that metformin reduced the viability of HO-8910 cells in a time- and concentration-dependent manner and induced cell apoptosis in a concentration-dependent manner. Metformin combined with DDP evidently inhibited cell viability and induced apoptosis. In addition, ERK1/2 and genes associated with apoptosis regulation, such as VEGF, VEGFR2, Bcl-2, Bax and caspase-3, exhibited differential expression in the HO-8910 cells. The present study demonstrated that expression of p-ERK1/2, VEGF, VEGFR2 and Bcl-2 was downregulated by treatment with increasing concentrations of metformin, whereas expression of Bax and caspase-3 was evidently upregulated. Taken together, these data demonstrate that metformin in combination with DDP reduces cell viability and induces apoptosis of human ovarian cancer cells.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA