Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Front Microbiol ; 15: 1341599, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38596371

RESUMO

Enteroviruses (EVs) are the main cause of a number of neurological diseases. Growing evidence has revealed that successful infection with enteroviruses is highly dependent on the host machinery, therefore, host proteins play a pivotal role in viral infections. Both host and viral proteins can undergo post-translational modification (PTM) which can regulate protein activity, stability, solubility and interactions with other proteins; thereby influencing various biological processes, including cell metabolism, metabolic, signaling pathways, cell death, and cancer development. During viral infection, both host and viral proteins regulate the viral life cycle through various PTMs and different mechanisms, including the regulation of host cell entry, viral protein synthesis, genome replication, and the antiviral immune response. Therefore, protein PTMs play important roles in EV infections. Here, we review the role of various host- and virus-associated PTMs during enterovirus infection.

2.
Int. j. morphol ; 41(1): 308-318, feb. 2023. ilus, tab, graf
Artigo em Inglês | LILACS | ID: biblio-1430503

RESUMO

SUMMARY: Gastrin plays a vital role in the development and progression of gastric cancer (GC). Its expression is up-regulated in GC tissues and several GC cell lines. Yet, the underlying mechanism remains to be investigated. Here, we aim to investigate the role and mechanism of gastrin in GC proliferation. Gastrin-overexpressing GC cell model was constructed using SGC7901 cells. Then the differentially expressed proteins were identified by iTRAQ analysis. Next, we use flow cytometry and immunofluorescence to study the effect of gastrin on the mitochondrial potential and mitochondria-derived ROS production. Finally, we studied the underlying mechanism of gastrin regulating mitochondrial function using Co-IP, mass spectrometry and immunofluorescence. Overexpression of gastrin promoted GC cell proliferation in vitro and in vivo. A total of 173 proteins were expressed differently between the controls and gastrin- overexpression cells and most of these proteins were involved in tumorigenesis and cell proliferation. Among them, Cox17, Cox5B and ATP5J that were all localized to the mitochondrial respiratory chain were down-regulated in gastrin-overexpression cells. Furthermore, gastrin overexpression led to mitochondrial potential decrease and mitochondria-derived ROS increase. Additionally, gastrin-induced ROS generation resulted in the inhibition of cell apoptosis via activating NF-kB, inhibiting Bax expression and promoting Bcl-2 expression. Finally, we found gastrin interacted with mitochondrial membrane protein Annexin A2 using Co-IP and mass spectrometry. Overexpr ession of gastrin inhibits GC cell apoptosis by inducing mitochondrial dysfunction through interacting with mitochondrial protein Annexin A2, then up-regulating ROS production to activate NF-kB and further leading to Bax/Bcl-2 ratio decrease.


La gastrina juega un papel vital en el desarrollo y progresión del cáncer gástrico (CG). Su expresión está regulada al alza en tejidos de CG y en varias líneas celulares de CG. Sin embargo, el mecanismo subyacente aun no se ha investigado. El objetivo de este estudio fue investigar el papel y el mecanismo de la gastrina en la proliferación de CG. El modelo de células CG que sobre expresan gastrina se construyó usando células SGC7901. Luego, las proteínas expresadas diferencialmente se identificaron mediante análisis iTRAQ. A continuación, utilizamos la citometría de flujo y la inmunofluorescencia para estudiar el efecto de la gastrina en el potencial mitocondrial y la producción de ROS derivada de las mitocondrias. Finalmente, estudiamos el mecanismo subyacente de la gastrina que regula la función mitocondrial utilizando Co-IP, espectrometría de masas e inmunofluorescencia. La sobreexpresión de gastrina promovió la proliferación de células CG in vitro e in vivo. Un total de 173 proteínas se expresaron de manera diferente entre los controles y las células con sobreexpresión de gastrina y la mayoría de estas proteínas estaban implicadas en la tumorigenesis y la proliferación celular. Entre estas, Cox17, Cox5B y ATP5J, todas localizadas en la cadena respiratoria mitocondrial, estaban reguladas a la baja en las células con sobreexpresión de gastrina. Además, la sobreexpresión de gastrina provocó una disminución del potencial mitocondrial y un aumento de las ROS derivadas de las mitocondrias. Por otra parte, la generación de ROS inducida por gastrina resultó en la inhibición de la apoptosis celular mediante la activación de NF-kB, inhibiendo la expresión de Bax y promoviendo la expresión de Bcl-2. Finalmente, encontramos que la gastrina interactuaba con la proteína de membrana mitocondrial Anexina A2 usando Co-IP y espectrometría de masas. La sobreexpresión de gastrina inhibe la apoptosis de las células CG al inducir la disfunción mitocondrial a través de la interacción con la proteína mitocondrial Anexina A2, luego regula el aumento de la producción de ROS para activar NF-kB y conduce aún más a la disminución de la relación Bax/Bcl-2.


Assuntos
Animais , Camundongos , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Gastrinas/metabolismo , Anexina A2/metabolismo , Mitocôndrias/patologia , Espectrometria de Massas , NF-kappa B , Imunofluorescência , Espécies Reativas de Oxigênio , Apoptose , Linhagem Celular Tumoral , Imunoprecipitação , Proliferação de Células , Carcinogênese , Citometria de Fluxo
3.
Transl Cancer Res ; 11(5): 1195-1206, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35706803

RESUMO

Background: The purpose of this study was to investigate the mechanism of action of muscone on breast cancer using network pharmacology and molecular docking techniques. Methods: Targets of muscone acid action were collected using the PubChem and SwissTargetPrediction databases. Relevant target sets of breast cancer were collected using the GeneCards database, and the intersection of the drug-disease targets was used as the potential target of muscone action in breast cancer. The STRING database was used to construct a target protein-protein interaction (PPI) network, and the data were imported into Cytoscape 3.7.1 for topological network analysis to obtain the core target genes of muscone in breast cancer. Gene Ontology (GO) functional enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed using the DAVID database. The correlation of core gene expression with breast cancer survival was analyzed using the online Kaplan-Meier plotter tool. Molecular docking of core target genes to muscone was performed using AutoDock Vina. Results: A total of 18 common targets of muscone and breast cancer were obtained through target intersection. The PPI map and topology analysis revealed that androgen receptor (AR), progesterone receptor (PGR), matrix metalloproteinase 9 (MMP9), prostaglandin-endoperoxide synthase 2 (PTGS2), heat shock protein 90 alpha family class A member 1 (HSP90AA1), mitogen-activated protein kinase 14 (MAPK14), and cytochrome P450 family 19 subfamily A member 1 (CYP19A1) might be the key targets of muscone acting on breast cancer. The GO enrichment analysis identified 60 terms, while the KEGG pathway enrichment analysis identified 7 signaling pathways, including steroid hormone biosynthesis, ovarian steroidogenesis, cancer pathways, and the tumor necrosis factor (TNF) signaling pathway. The results of survival stage analysis showed that the binding activity between muskone and key targets was better than other targets. The molecular docking results showed that muscone had the highest docking affinity for the key target CYP19A1 gene at -7.0 kJ/moL. Conclusions: Muscone might exert anti-breast cancer effects through cancer pathways, ovarian steroidogenesis, and TNF signaling pathways and has the potential to be developed as a clinical agent.

4.
Biomed Res Int ; 2021: 5590037, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33937399

RESUMO

Overexpressed gastrin is reported to promote oncogenesis and development of gastric cancer by inhibiting apoptosis of cancer cells; however, the underlying mechanism remains unclear. Our study is aimed at revealing the mechanism underlying the effect of gastrin on apoptosis of gastric cancer cells. Gastrin-interfering cell line was constructed by stably transfecting gastrin-specific pshRNA plasmid to gastric cancer cell line BGC-823. Then, differentially expressed proteins between untreated BGC-823 and gastrin-interfering BGC-823 cell lines were detected by the iTRAQ technique. GO and KEGG analysis was used to analyze the differentially expressed genes that code these differentially expressed proteins. The Annexin V-FITC staining assay was used to detect gastric cancer cell apoptosis. The DCFH-DA fluorescent probe staining assay was used to measure intracellular ROS. Mitochondrial membrane potential was detected by flow cytometry. Western blot was used to analyze the mitochondria respiratory chain proteins and apoptosis-related proteins. A total of 107 differentially expressed proteins were identified by iTRAQ. GO and KEGG analysis showed that proteins coded by the corresponding differentially expressed genes were mainly enriched in the mitochondrial oxidative respiratory chain, and the expression of three proteins (COX17, COX5B, ATP5J) was upregulated. The three proteins with higher scores were verified by Western blot. The apoptosis rate of the gastrin knockdown cancer cell was significantly increased; meanwhile, gastrin knockdown leads to increase of membrane potential and decrease of intracellular ROS production. Additionally, Bax was significantly increased, whereas NF-κB-p65 and Bcl-2 were downregulated after knockdown of gastrin. Concomitantly, pretreatment with NAC reversed the effect of gastrin on the Bax and Bcl-2 expression. Gastrin promotes the production of ROS from mitochondria, activates NF-κB, and inhibits apoptosis via modulating the expression level of Bcl-2 and Bax.


Assuntos
Apoptose/genética , Gastrinas , Espécies Reativas de Oxigênio , Neoplasias Gástricas , Linhagem Celular Tumoral , Gastrinas/genética , Gastrinas/metabolismo , Humanos , Mitocôndrias/metabolismo , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/análise , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo
5.
Chin J Integr Med ; 27(7): 514-519, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32144561

RESUMO

OBJECTIVE: To study the effect and mechanism of Huayu Wan (, HYW) in combination of chemotherapy of tumor treatment. METHODS: HYW serum was added in Lewis cells to assess its impact on fluorescent doxorubicin delivery in vitro. Then, Lewis tumor cells was implanted in C57BL/6 mice via xenograft transplantation. Tumor growth was measured and signal intensity corresponding to blood flow was assessed by laser doppler perfusion imaging (LDPI). Finally, the effect of HYW on the effificacy of doxorubicin was studied. RESULTS: HYW can improve the transfer of fluorescent doxorubicin into cells. The blood flow signal in the tumor tissues of the HYW group was higher than that of the control group (P<0.01). Furthermore, HYW improved drug delivery of doxorubicin to tumor tissues, and this activity was associated with HYW-induced microvascular proliferation (P<0.01). CONCLUSIONS: HYW can promote microangiogenesis and increase blood supply in tumor tissues, which in turn may increase the risk of metastasis. At the same time, HYW increases drug delivery and improves the effificacy of chemotherapy drugs through vascular proliferation. Therefore, rational judgment must be exercised when considering applying HYW to an antitumor regimen.


Assuntos
Doxorrubicina , Neoplasias Pulmonares , Animais , Linhagem Celular Tumoral , Doxorrubicina/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Int J Mol Med ; 42(2): 811-820, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29749427

RESUMO

Fisetin, a natural flavonoid found in a variety of edible and medical plants, has been suggested to inhibit the proliferation of various tumor cells and to induce apoptosis. However, the effects of fisetin on breast cancer have rarely been reported and the underlying mechanism is still undefined. The present study explored the anti­cancer effects of fisetin on mammary carcinoma cells and the underlying mechanisms. Following treatment with fisetin, viability of 4T1, MCF­7 and MDA­MB­231 cells were measured by MTT assay. The inhibitory effects of fisetin on proliferation, migration and invasion were evaluated in 4T1 cells using proliferation array, wound­healing assay, and HUV­EC­C­cell barrier based on electrical cell­substrate impedance sensing platform. Cell apoptosis was analyzed by flow cytometry, and western blotting analysis was performed to identify target molecules. A 4T1 orthotopic mammary tumor model was used to assess the fisetin­inhibition on tumor growth in vivo. Test kits were used to examine the liver and kidney function of tumor­bearing mice. The results suggest that fisetin suppressed the proliferation of breast cancer cells, suppressed the metastasis and invasiveness of 4T1 cells, and induced the apoptosis of 4T1 cells in vitro. The potent anti­cancer effect of fisetin was associated with the regulation of the phosphatidylinositol­3­kinase/protein kinase B/mammalian target of rapamycin pathway. In vivo experiments demonstrated that fisetin suppressed the growth of 4T1 cell­derived orthotopic breast tumors and enhanced tumor cell apoptosis, and the evaluated alanine amino transferase and aspartate amino transferase levels in serum of tumor­bearing mice suggested that fisetin may lead to side effects on liver biochemical function. The present study confirms that fisetin exerted an anti­mammary carcinoma effect. However, in vivo experiments also revealed that fisetin had low solubility and low bioavailability. Further investigation is required to determine the clinical value of fisetin.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Flavonoides/uso terapêutico , Neoplasias Mamárias Animais/tratamento farmacológico , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Flavonoides/farmacologia , Flavonóis , Células Endoteliais da Veia Umbilical Humana , Humanos , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais/efeitos dos fármacos
7.
PLoS One ; 12(7): e0181286, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28686699

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0174095.].

8.
PLoS One ; 12(3): e0174095, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28328929

RESUMO

OBJECTIVES: To evaluate whether urinary iodine concentration (UIC) can predict goiter among school-age children, and to assess the association between UIC and goiter prevalence. METHODS: We searched the MEDLINE, EMBASE, Cochrane Library (Cochrane Database of Systematic Reviews), Web of Science, CNKI, VIP, and Wan Fang databases for relevant reports in both English and Chinese up to August 25, 2016. The mean differences (MD) and 95% confidence intervals (CI) were calculated for the UIC and goiter prevalence assessments. Pooled odds ratios and 95% CIs were used to compare the prevalences of goiter in the different UIC groups. RESULTS: We identified 11 case-control studies, and found that children with goiter had lower UIC values, compared to children without goiter (MD: -1.82, 95% CI: -3.24, -0.40, p < 0.05). An increased risk of goiter was associated with UIC values of < 20 µg/L or > 200 µg/L. CONCLUSION: The results of our meta-analysis suggest that lower UIC values were associated with an increased risk of goiter, and that iodine deficiency may lead to an increased risk of goiter. Furthermore, we observed U-shaped relationships between UIC and the prevalence of goiter, which suggests that both severe iodine deficiency and excessive iodine intake may lead to increased risks of goiter.


Assuntos
Biomarcadores/urina , Bócio/diagnóstico , Bócio/urina , Iodo/urina , Adolescente , Estudos de Casos e Controles , Criança , Feminino , Bócio/epidemiologia , Humanos , Masculino , Prevalência , Doenças da Glândula Tireoide/diagnóstico , Doenças da Glândula Tireoide/epidemiologia , Doenças da Glândula Tireoide/urina , Glândula Tireoide/metabolismo
9.
Medicine (Baltimore) ; 96(47): e8487, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29381923

RESUMO

Colon cancer arises from the accumulations of genetic and epigenetic changes. Currently, profiles of DNA methylation and gene expression of colon cancer have not been elucidated clearly. This articles aims to characterize the profile of DNA methylation and gene expression of colon cancer systemically, and acquire candidate genes potentially regulated by altered methylation for this disease.Data were downloaded from The Cancer Genome Atlas database. Differentially methylated CpG sites (DMCs) and differentially methylated regions (DMRs) were calculated via COHCAP. Differentially expressed genes (DEGs) were identified by DESeq2. Weighted gene co-expression network analysis (WGCNA) package in R was applied for WGCNA.Data of 275 solid tumor tissues and 19 adjacent tumor tissues of colon cancer were obtained. A total of 1828 DMCs, including 1390 hypermethylated and 438 hypomethylated CpG sites, were identified between tumor and normal groups. A total of 789 DEGs, containing 435 upregulated genes and 354 downregulated genes were observed. It revealed that 8 DMRs-DEGs and 95 DMCs-DEGs pairs were significantly correlated. Furthermore, genes of yellow and brown modules from WGCNA were significantly correlated with tumor/normal status, and significantly enriched in peroxisome proliferator activated receptor signaling pathway, glutamatergic synapse, and neuroactive ligand-receptor interaction. Genes in the above 2 modules were also significantly enriched in DMCs or DMRs-associated genes. Specifically, ADHFE1, HAND2, and GNAO1 were hypermethylated and downregulated in colon cancer, suggesting that the low expression levels of these genes may be regulated by DNA hypermethylation. In addition, the 3 genes were involved in brown module of WGCNA, indicating their important roles in colon cancer.The investigation of the relationship between DNA methylation and gene expression may help to understand the effect of DNA methylation alteration on genes expression, especially gene co-expression network in the development of colon cancer. Genes such as ADHFE1, HAND2, and GNAO1 may be served as potential candidates for diagnosis and therapy targets in colon cancer.


Assuntos
Neoplasias do Colo/genética , Neoplasias do Colo/fisiopatologia , Metilação de DNA/fisiologia , Expressão Gênica/fisiologia , Oxirredutases do Álcool/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Ilhas de CpG/fisiologia , Bases de Dados Genéticas , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Redes Reguladoras de Genes/fisiologia , Humanos , Proteínas Mitocondriais/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Transdução de Sinais/fisiologia
10.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 34(6): 745-50, 2014 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-25046961

RESUMO

OBJECTIVE: To explore the effect of Lignum Sappan (LS) containing serum on the proliferation cycle arrest of human lung cancer cell line PG and its molecular mechanism. METHODS: The lung cancer PG cells were divided into four groups, i.e., the blank control group, the LS group, the LS plus cisplatin group, and the cisplatin group. They were cultured by RPMI-1640 with 20% blank serum, RPMI-1640 with 20% LS containing serum, RPMI-1640 with 20% LS containing serum plus 1 microg/mL cisplatin, and RPMI-1640 with 20% blank serum plus 1 microg/mL cisplatin, respectively. The morphology of PG cells was observed using light microscope and laser scanning confocal microscope in each group. The cell cycle arrest was observed using flow cytometry. The expression of P16 and Rb1 mRNA was tested by PCR method. RESULTS: Under the light microscope and laser scanning confocal microscope, the apoptosis degree of PG cells in the LS group was significant, but less than that of the LS plus cisplatin group as well as the cisplatin group. Compared with the blank control group, the proportion of PG cells increased at G0/ G1 and S phases (P < 0.05) and decreased at G2/M phase (P < 0.01) in the LS group; The proportion of PG cells increased at G2/M and S phases (P < 0.05, P < 0.01) and decreased at G0/G1 phase (P < 0.01) in the LS plus cisplatin group as well as the cisplatin group. Compared with the LS group, the proportion of PG cells increased at G2/M and S phases (P < 0.05, P < 0.01) and decreased at G0/G1 phase (P < 0.01) in the LS plus cisplatin group as well as the cisplatin group. There was no statistical difference in PG cells at each phase between the cisplatin group and the LS plus cisplatin group (P > 0.05). The expression of P16 and Rb1 mRNA increased in the LS group, when compared with the blank control group. They also increased in the cisplatin group and the LS plus cisplatin group, higher than that of the LS group (P < 0.05). There was no statistical difference in the expression of P16 and Rb1 mRNA between the cisplatin group and the LS plus cisplatin group (P > 0.05). CONCLUSION: LS containing serum induced PG cell apoptosis by up-regulating the mRNA transcription levels of P16 and Rb1, thus resulting in PG cell arrest at G0/G1 and S phases, which was different from the manner of cisplatin (achieved by arresting PG cells at G2/M and S phases through regulating cyclinB1 mRNA transcription).


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Neoplasias Pulmonares/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...