Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biol Direct ; 18(1): 19, 2023 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-37088822

RESUMO

BACKGROUND: Colorectal cancer (CRC) is one of the most common malignant tumors of the digestive system, ranking third for morbidity and mortality worldwide. At present, no effective control method is available for this cancer type. In tumor cells, especially iron metabolization, is necessary for its growth and proliferation. High levels of iron are an important feature to maintain tumor growth; however, the overall mechanism remains unclear. METHODS: We used western blotting, immunohistochemistry (IHC) and real-time quantitative PCR to analyze the expression of IGF2BP2 in cell lines and tissues. Further, RNA-sequencing, RNA immunoprecipitation and methylated RNA immunoprecipitation experiments explored the specific binding of target genes. Moreover, the RNA stability assay was performed to determine the half-life of genes downstream of IGF2BP2. In addition, the Cell Counting Kit-8, colony formation assay, 5-ethynyl-2'-deoxyuridine assay and flow cytometry were used to evaluate the effects of IGF2BP2 on proliferation and iron metabolism. Lastly, the role of IGF2BP2 in promoting CRC growth was demonstrated in animal models. RESULTS: We observed that IGF2BP2 is associated with iron homeostasis and that TFRC is a downstream target of IGF2BP2. Further, overexpression of TFRC can rescue the growth of IGF2BP2-knockdown CRC cells. Mechanistically, we determined that IGF2BP2 regulates TFRC methylation via METTL4, thereby regulating iron metabolism and promoting CRC growth. Furthermore, using animal models, we observed that IGF2BP2 promotes CRC growth. CONCLUSION: IGF2BP2 regulates TFRC mRNA methylation via METTL4, thereby regulating iron metabolism and promoting CRC growth. Our study highlights the key roles of IGF2BP2 in CRC carcinogenesis and the iron transport pathways.


Assuntos
Neoplasias Colorretais , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Proliferação de Células/genética , Carcinogênese/genética , RNA , Regulação Neoplásica da Expressão Gênica
2.
Mil Med Res ; 9(1): 58, 2022 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-36229865

RESUMO

BACKGROUND: Abnormal myocardial Nav1.5 expression and function cause lethal ventricular arrhythmias during myocardial ischemia-reperfusion (I/R). Protein inhibitor of activated STAT Y (PIASy)-mediated caveolin-3 (Cav-3) SUMO modification affects Cav-3 binding to the voltage-gated sodium channel 1.5 (Nav1.5). PIASy activity is increased after myocardial I/R, but it is unclear whether this is attributable to plasma membrane Nav1.5 downregulation and ventricular arrhythmias. METHODS: Using recombinant adeno-associated virus subtype 9 (AAV9), rat cardiac PIASy was silenced using intraventricular injection of PIASy short hairpin RNA (shRNA). After two weeks, rat hearts were subjected to I/R and electrocardiography was performed to assess malignant arrhythmias. Tissues from peri-infarct areas of the left ventricle were collected for molecular biological measurements. RESULTS: PIASy was upregulated by I/R (P < 0.01), with increased SUMO2/3 modification of Cav-3 and reduced membrane Nav1.5 density (P < 0.01). AAV9-PIASy shRNA intraventricular injection into the rat heart downregulated PIASy after I/R, at both mRNA and protein levels (P < 0.05 vs. Scramble-shRNA + I/R group), decreased SUMO-modified Cav-3 levels, enhanced Cav-3 binding to Nav1.5, and prevented I/R-induced decrease of Nav1.5 and Cav-3 co-localization in the intercalated disc and lateral membrane. PIASy silencing in rat hearts reduced I/R-induced fatal arrhythmias, which was reflected by a modest decrease in the duration of ventricular fibrillation (VF; P < 0.05 vs. Scramble-shRNA + I/R group) and a significantly reduced arrhythmia score (P < 0.01 vs. Scramble-shRNA + I/R group). The anti-arrhythmic effects of PIASy silencing were also evidenced by decreased episodes of ventricular tachycardia (VT), sustained VT and VF, especially at the time 5-10 min after ischemia (P < 0.05 vs. Scramble-shRNA + IR group). Using in vitro human embryonic kidney 293 T (HEK293T) cells and isolated adult rat cardiomyocyte models exposed to hypoxia/reoxygenation (H/R), we confirmed that increased PIASy promoted Cav-3 modification by SUMO2/3 and Nav1.5/Cav-3 dissociation after H/R. Mutation of SUMO consensus lysine sites in Cav-3 (K38R or K144R) altered the membrane expression levels of Nav1.5 and Cav-3 before and after H/R in HEK293T cells. CONCLUSIONS: I/R-induced cardiac PIASy activation increased Cav-3 SUMOylation by SUMO2/3 and dysregulated Nav1.5-related ventricular arrhythmias. Cardiac-targeted PIASy silencing mediated Cav-3 deSUMOylation and partially prevented I/R-induced Nav1.5 downregulation in the plasma membrane of cardiomyocytes, and subsequent ventricular arrhythmias in rats. PIASy was identified as a potential therapeutic target for life-threatening arrhythmias in patients with ischemic heart diseases.


Assuntos
Antiarrítmicos , Caveolina 3 , Proteínas de Ligação a Poli-ADP-Ribose/genética , Proteínas Inibidoras de STAT Ativados/genética , Animais , Arritmias Cardíacas/genética , Caveolina 3/genética , Caveolina 3/metabolismo , Regulação para Baixo , Inativação Gênica , Células HEK293 , Humanos , Isquemia/complicações , Lisina/genética , Lisina/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , RNA Mensageiro , RNA Interferente Pequeno , Ratos , Reperfusão/efeitos adversos
3.
Curr Med Sci ; 38(6): 997-1004, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30536061

RESUMO

The aim of the present study was to explore the effects of annexin A1 (ANXA1) mimetic peptide AC2-26 on sepsis-induced cardiomyocyte apoptosis in vivo and in vitro and the underlying mechanisms. In the in vivo study, a rat septic model was established by the cecal ligation and puncture (CLP). The rats were divided into control group, sepsis group and AC2-26 group. The rats in the AC2-26 group were intraperitoneally injected with AC2-26 (1 mg/kg) 2 h before CLP, and those in the control group and sepsis group were injected with the same volume of normal saline. The myocardial tissue was examined by hematoxylin and eosin (HE) staining and transmission electron microscopy (TEM). Furthermore, myocardial apoptosis was measured by terminal dUTP nick end-labeling (TUNEL) assay. In the in vitro study, H9C2 cells were cultured and divided into three groups: control group, in which cells were only given the basic culture medium; LPS group, in which cells were treated with 10 µg/mL LPS; AC2-26 group, in which cells were treated with 0.5 µmol/L AC2-26 2 h before 10 µg/mL LPS was given. The apoptosis of H9C2 cells was detected by flow cytometry. The levels of lipoxin A4 receptor (LXA4), phosphoinositide-3-kinase (PI3K) and protein kinase B (PKB or AKT) protein were measured by Western blotting, the activity of NF-κB and the level of TNF-α by ELISA and the activities of caspase-3/8 by using the caspase activity kits. The in vivo study showed that the myocardial pathological damage and myocardial ultrastructural damage were significantly alleviated and the myocardial apoptosis significantly decreased in the AC2-26 group as compared with the sepsis group (P<0.05 for all). The in vivo study revealed that the apoptosis of H9C2 cells was profoundly ameliorated in the AC2-26 group relative to the sepsis group (P<0.05). The protein expression levels of LXA4 were significantly up-regulated, and those of PI3K and AKT prominently down-regulated in the AC2-26 group when compared with those in the LPS group (P<0.05 for all). The activity of NF-κB was greatly inhibited and the level of TNF-α markedly decreased in the AC2-26 group as compared with those in the LPS group (P<0.05 for all). AC2-26 treatment also significantly suppressed the activities of caspase-3/8 in H9C2 cells. In conclusion, these findings suggest that AC2-26 may alleviate the sepsis-induced cardiomyocyte apoptosis in vivo and in vivo through the LXA4/PI3K/AKT signaling pathway.


Assuntos
Anexina A1/metabolismo , Apoptose/fisiologia , Miócitos Cardíacos/metabolismo , Peptídeos/metabolismo , Sepse/metabolismo , Transdução de Sinais/fisiologia , Animais , Regulação para Baixo/fisiologia , Lipoxinas/metabolismo , Masculino , Miocárdio/metabolismo , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA