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.
Cell Mol Life Sci ; 81(1): 387, 2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39249529

RESUMO

BACKGROUND: Dysregulated lipid oxidation occurs in several pathological processes characterized by cell proliferation and migration. Nonetheless, the molecular mechanism of lipid oxidation is not well appreciated in liver fibrosis, which is accompanied by enhanced fibroblast proliferation and migration. METHODS: We investigated the causes and consequences of lipid oxidation in liver fibrosis using cultured cells, animal models, and clinical samples. RESULTS: Increased ecto-nucleotide pyrophosphatase/phosphodiesterase (ENPP1) expression caused increased lipid oxidation, resulting in the proliferation and migration of hepatic stellate cells (HSCs) that lead to liver fibrosis, whereas fibroblast-specific ENPP1 knockout reversing these results. Elevated ENPP1 and N6-methyladenosine (m6A) levels were associated with high expression of Wilms tumor 1 associated protein (WTAP). Mechanistically, WTAP-mediated m6A methylation of the 3'UTR of ENPP1 mRNA and induces its translation dependent of YTH domain family proteins 1 (YTHDF1). Additionally, ENPP1 could interact with hypoxia inducible lipid droplet associated (HILPDA) directly; overexpression of ENPP1 further recruits HILPDA-mediated lipid oxidation, thereby promotes HSCs proliferation and migration, while inhibition of ENPP1 expression produced the opposite effect. Clinically, increased expression of WTAP, YTHDF1, ENPP1, and HILPDA, and increased m6A mRNA content, enhanced lipid oxidation, and increased collagen deposition in human liver fibrosis tissues. CONCLUSIONS: We describe a novel mechanism in which WTAP catalyzes m6A methylation of ENPP1 in a YTHDF1-dependent manner to enhance lipid oxidation, promoting HSCs proliferation and migration and liver fibrosis.


Assuntos
Adenosina , Proliferação de Células , Metabolismo dos Lipídeos , Cirrose Hepática , Oxirredução , Diester Fosfórico Hidrolases , Pirofosfatases , RNA Mensageiro , Pirofosfatases/metabolismo , Pirofosfatases/genética , Humanos , Diester Fosfórico Hidrolases/metabolismo , Diester Fosfórico Hidrolases/genética , Animais , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Cirrose Hepática/genética , Adenosina/análogos & derivados , Adenosina/metabolismo , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proliferação de Células/genética , Metabolismo dos Lipídeos/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Movimento Celular/genética , Camundongos Endogâmicos C57BL , Masculino , Epigênese Genética , Fibroblastos/metabolismo , Fibroblastos/patologia , Metilação , Fatores de Processamento de RNA , Proteínas de Ciclo Celular
2.
Cell Signal ; 122: 111302, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39025344

RESUMO

Liver fibrosis is a pathological process caused by a variety of chronic liver diseases. Currently, therapeutic options for liver fibrosis are very limited, highlighting the urgent need to explore new treatment approaches. Epigenetic modifications and epitranscriptomic modifications, as reversible regulatory mechanisms, are involved in the development of liver fibrosis. In recent years, researches in epitranscriptomics and epigenetics have opened new perspectives for understanding the pathogenesis of liver fibrosis. Exploring the epigenetic mechanisms of liver fibrosis may provide valuable insights into the development of new therapies for chronic liver diseases. This review primarily focus on the regulatory mechanisms of N6-methyladenosine (m6A) modification, non-coding RNA, and DNA methylation in organ fibrosis. It discusses the interactions between m6A modification and DNA methylation, as well as between m6A modification and non-coding RNA, providing a reference for understanding the interplay between epitranscriptomics and epigenetics.


Assuntos
Adenosina , Metilação de DNA , Epigênese Genética , Cirrose Hepática , RNA não Traduzido , Humanos , Cirrose Hepática/genética , Cirrose Hepática/patologia , Cirrose Hepática/metabolismo , RNA não Traduzido/metabolismo , RNA não Traduzido/genética , Adenosina/análogos & derivados , Adenosina/metabolismo , Animais , Transcriptoma
3.
Free Radic Biol Med ; 218: 166-177, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38582229

RESUMO

BACKGROUND: Dysregulated ecto-nucleotide pyrophosphatase/phosphodiesterase (ENPP) family occurs in metabolic reprogramming pathological processes. Nonetheless, the epigenetic mechanisms by which ENPP family impacts NAFLD, also known as metabolic dysfunction-associated steatotic liver disease (MASLD), is poorly appreciated. METHODS: We investigated the causes and consequences of ENPP1 promoter hypomethylation may boost NAFLD using NAFLD clinical samples, as well as revealed the underlying mechanisms using high-fat diet (HFD) + carbon tetrachloride (CCl4) induced mouse model of NAFLD and FFA treatment of cultured hepatocyte. RESULTS: Herein, we report that the expression level of ENPP1 are increased in patients with NAFLD liver tissue and in mouse model of NAFLD. Hypomethylation of ENPP1, is associated with the perpetuation of hepatocyte autophagy and liver fibrosis in the NAFLD. ENPP1 hypomethylation is mediated by the DNA demethylase TET3 in NAFLD liver fibrosis and hepatocyte autophagy. Additionally, knockdown of TET3 methylated ENPP1 promoter, reduced the ENPP1 expression, ameliorated the experimental NAFLD. Mechanistically, TET3 epigenetically promoted ENPP1 expression via hypomethylation of the promoter. Knocking down TET3 can inhibit the hepatocyte autophagy but an overexpression of ENPP1 showing rescue effect. CONCLUSIONS: We describe a novel epigenetic mechanism wherein TET3 promoted ENPP1 expression through promoter hypomethylation is a critical mediator of NAFLD. Our findings provide new insight into the development of preventative measures for NAFLD.


Assuntos
Autofagia , Metilação de DNA , Dioxigenases , Modelos Animais de Doenças , Epigênese Genética , Hepatócitos , Hepatopatia Gordurosa não Alcoólica , Diester Fosfórico Hidrolases , Regiões Promotoras Genéticas , Pirofosfatases , Animais , Humanos , Masculino , Camundongos , Autofagia/genética , Tetracloreto de Carbono/toxicidade , Dieta Hiperlipídica/efeitos adversos , Dioxigenases/genética , Dioxigenases/metabolismo , Hepatócitos/metabolismo , Hepatócitos/patologia , Cirrose Hepática/genética , Cirrose Hepática/patologia , Cirrose Hepática/metabolismo , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Diester Fosfórico Hidrolases/genética , Diester Fosfórico Hidrolases/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Pirofosfatases/genética , Pirofosfatases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA