1
|
Shen X, Shi C, Xu J, Zhi F, Luo K, Di Y, Li W, Ma W, Jiang Y, Sun H. Intestinal microbiota homeostasis analysis in riboflavin-treated alcoholic liver disease. Commun Biol 2024; 7:1030. [PMID: 39169207 PMCID: PMC11339332 DOI: 10.1038/s42003-024-06722-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 08/12/2024] [Indexed: 08/23/2024] Open
Abstract
Alcoholic liver disease (ALD) is a disease with high incidence, limited therapies, and poor prognosis. The present study aims to investigate the effect of riboflavin on ALD and explore its potential therapeutic mechanisms. C57BL/6 mice were divided into the control, alcohol, and alcohol+ riboflavin groups. 16S rRNA-seq and RNA-seq analysis were utilized to analyze the polymorphism of intestinal microbiota and the transcriptome heterogeneity respectively. KEGG and GO enrichment analysis were performed. CIBERSORTx was applied to evaluate the immune cell infiltration level. Publicly available transcriptome data of ALD was enrolled and combined with the RNA-seq data to identify the immune subtypes of ALD. Pathological and histology analysis demonstrated that riboflavin reversed the progression of ALD. 16S rRNA-seq results showed that riboflavin could regulate alcohol-induced intestinal microbiota alteration. Intestinal microbiota polymorphism analysis indicated that VLIDP may contribute to the progression of ALD. Based on the VLIDP pathway, two subtypes were identified. Immune microenvironment analysis indicated that the upregulated inflammatory factors may be important regulators of ALD. In conclusion, intestinal microbiota homeostasis was associated with the protective effect of riboflavin against ALD, which was likely mediated by modulating inflammatory cell infiltration. Riboflavin emerges as a promising therapeutic candidate for the management of ALD.
Collapse
Affiliation(s)
- Xiuyun Shen
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, PR China
| | - Chunpeng Shi
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Jincheng Xu
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Fengnan Zhi
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Kunpeng Luo
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yuzhu Di
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Wanhong Li
- Pharmaceutical Experiment Teaching Center College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Wanjing Ma
- Pharmaceutical Experiment Teaching Center College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yanan Jiang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, PR China.
| | - Hui Sun
- Pharmaceutical Experiment Teaching Center College of Pharmacy, Harbin Medical University, Harbin, PR China.
| |
Collapse
|
2
|
Zheng J, Li Z, Xu H. Intestinal Microbiotas and Alcoholic Hepatitis: Pathogenesis and Therapeutic Value. Int J Mol Sci 2023; 24:14809. [PMID: 37834256 PMCID: PMC10573193 DOI: 10.3390/ijms241914809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/21/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Alcoholic hepatitis (AH) is a rapidly progressing and severe stage of alcoholic liver disease, presenting a grim prognosis. Extensive research has elucidated several underlying mechanisms that contribute to the development of AH, including metabolic alterations, immune stimulation, and intestinal dysbiosis. These pathological changes intricately intertwine during the progression of AH. Notably, recent studies have increasingly highlighted the pivotal role of alterations in the intestinal microbiota in the pathogenesis of AH. Consequently, future investigations should place significant emphasis on exploring the dynamics of intestinal microbiota. In this comprehensive review, we consolidate the primary causes of AH while underscoring the influence of gut microbes. Furthermore, by examining AH treatment strategies, we delineate the potential therapeutic value of interventions targeting the gut microbiota. Given the existing limitations in AH treatment options, we anticipate that this review will contribute to forthcoming research endeavors aimed at advancing AH treatment modalities.
Collapse
Affiliation(s)
- Jiazhen Zheng
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (J.Z.); (Z.L.)
| | - Ziyi Li
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (J.Z.); (Z.L.)
| | - Hengyi Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| |
Collapse
|
3
|
Li WH, Zhang L, Li YY, Wang XY, Li JL, Zhao SN, Ni MQ, Li Q, Sun H. Apolipoprotein A-IV Has Bi-Functional Actions in Alcoholic Hepatitis by Regulating Hepatocyte Injury and Immune Cell Infiltration. Int J Mol Sci 2022; 24:ijms24010670. [PMID: 36614113 PMCID: PMC9820766 DOI: 10.3390/ijms24010670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023] Open
Abstract
Alcohol abuse can lead to alcoholic hepatitis (AH), a worldwide public health issue with high morbidity and mortality. Here, we identified apolipoprotein A-IV (APOA4) as a biomarker and potential therapeutic target for AH. APOA4 expression was detected by Gene Expression Omnibus (GEO) databases, Immunohistochemistry, and qRT-PCR in AH. Bioinformatics Methods (protein-protein interaction (PPI) network, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and Gene Set Enrichment Analysis (GSEA) were used to show down-stream gene and pathways of APOA4 in AH. AML-12 cells were used to evaluate the biological function of APOA4 using an ELISA kit (AST, ALT, and IL-1β) and flow cytometry (ROS activity). Both in vivo and in vitro, APOA4 expression was significantly elevated in the AH model induced by alcohol (ETOH). AML-12 cell damage was specifically repaired by APOA4 deficiency, while AST, ALT, and IL-1β activity that was increased by ETOH (200 µmol, 12 h) were suppressed. APOA4 inhibition increased intracellular ROS induced by ETOH, which was detected by flow cytometry. Functional and PPI network analyses showed Fcgamma receptor (FCGR) and platelet activation signaling were potential downstream pathways. We identified CIDEC as a downstream gene of APOA4. The CIDEC AUC values for the ROC curves were 0.861. At the same time, APOA4 silencing downregulated the expression of CIDEC, whereas the knockdown of CIDEC did not influence the expression of APOA4 in AML-12 cells. Collectively, APOA4 regulates CIDEC expression and immune cell infiltration and may hold great potential as a biomarker and therapeutic target for AH.
Collapse
Affiliation(s)
- Wan-Hong Li
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Li Zhang
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yue-Ying Li
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Xin-Yue Wang
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Jin-Liang Li
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Shu-Ning Zhao
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Ming-Qi Ni
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Qian Li
- Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Correspondence: (Q.L.); (H.S.); Tel./Fax: +86-451-86699347 (Q.L.)
| | - Hui Sun
- Pharmaceutical Experiment Teaching Center, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Correspondence: (Q.L.); (H.S.); Tel./Fax: +86-451-86699347 (Q.L.)
| |
Collapse
|
4
|
Systemic pharmacology understanding of the key mechanism of Sedum sarmentosum Bunge in treating hepatitis. Naunyn Schmiedebergs Arch Pharmacol 2020; 394:421-430. [PMID: 32734365 DOI: 10.1007/s00210-020-01952-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022]
Abstract
Sedum sarmentosum Bunge is a Traditional Chinese Medicine that is widely used in treating hepatitis, whereas the detailed mechanisms have not been fully interpreted. A systemic pharmacology method including absorption, distribution, metabolism and elimination screening, drug targeting, interaction network plotting, and enrichment analysis was applied for exploring the underlying mechanisms of Sedum sarmentosum Bunge in the treatment of hepatitis. A total of 47 ingredients were identified in Sedum sarmentosum Bunge, and 5 active ingredients (DFV, isorhamnetin, beta-sitosterol, luteolin and quercetin) were screened out with the criteria of oral bioavailability (OB) ≥ 30% and drug-likeness (DL) ≥ 0.18. Those 5 ingredients interacted with 170 targets, 163 of which were hepatitis-related. By compound-target-disease network plotting, protein-protein interaction network plotting and enrichment analysis, the pathways that the 5 ingredients engaged in during hepatitis development and progression were investigated, such as threonine-protein kinase signaling. The integrated systemic pharmacology analysis facilitates the in-depth understanding of Sedum sarmentosum Bunge in the hepatitis treatment, which also paves the way for further knowledge of the molecular mechanism of Sedum sarmentosum Bunge in treating hepatitis.
Collapse
|