Your browser doesn't support javascript.
loading
Identification of key genes increasing susceptibility to atrial fibrillation in nonalcoholic fatty liver disease and the potential mechanisms: mitochondrial dysfunction and systemic inflammation.
Zhong, Baiyin; Xie, Zhonghui; Zhang, Jianhong; Xie, Xing; Xie, Yuankang; Xie, Binhui; Wang, Jing; Liu, Chuanbin.
Afiliação
  • Zhong B; Department of Hepatobiliary Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China.
  • Xie Z; Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China.
  • Zhang J; Department of Hepatobiliary Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China.
  • Xie X; Department of Hepatobiliary Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China.
  • Xie Y; Department of Hepatobiliary Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China.
  • Xie B; Department of Hepatobiliary Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China.
  • Wang J; Department of General Medicine, The First Medical Center of PLA General Hospital, Beijing, China.
  • Liu C; Western Medical Branch of PLA General Hospital, Beijing, China.
Front Pharmacol ; 15: 1360974, 2024.
Article em En | MEDLINE | ID: mdl-38549670
ABSTRACT

Background:

Non-alcoholic fatty liver disease (NAFLD) and atrial fibrillation (AF) are major health burdens, with emerging evidence suggesting NAFLD as a significant risk factor for AF, but the mechanism is remain unclear.

Methods:

In this study, we analyzed gene expression data from NAFLD (GSE89632) and AF (GSE75092) datasets from the Gene Expression Omnibus. We identified co-upregulated and co-downregulated genes between NAFLD and AF, assessed diagnostic potential of specific genes, conducted immune infiltration analysis, and performed molecular docking studies with sodium glucose co-transporter 2 inhibitors (SGLT2i).

Results:

We identified eight co-upregulated and 31 co-downregulated genes between NAFLD and AF. Genes such as AMOT, PDE11A, TYMS, TMEM98, and PTGS2 demonstrated substantial diagnostic potential for identifying NAFLD patients at risk of AF. Immune infiltration analysis discovered an elevated presence of CD8 T cells, γδ T cells, and M2 macrophages in NAFLD livers, linking systemic inflammation to NAFLD and AF. Additionally, studies have shown that a connection between mitochondrial dysfunction and several hub genes like DGAT1, TYMS, and PTGS2, suggesting that mitochondrial disturbances may underpin the systemic inflammation in NAFLD, which possibly exacerbating AF. Molecular docking studies indicated that empagliflozin's binding affinity with key genes such as DGAT1, TYMS, and PTGS2 presents a novel therapeutic avenue for NAFLD-associated AF.

Conclusion:

Our study firstly discovered that AMOT, PDE11A, TYMS, TMEM98, and PTGS2 are associated with NAFLD-related AF and hold strong diagnostic values. Our study also indicates that mitochondrial dysfunction and systemic inflammation may be potential mechanisms bridging NAFLD and AF. Additionally, we identified empagliflozin as a potentially effective therapeutic agent for NAFLD-related AF at the molecular structure level. These novel insights contribute to the further understanding, diagnosis, and intervention of NAFLD-related AF.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: Front Pharmacol Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: Front Pharmacol Ano de publicação: 2024 Tipo de documento: Article