1.Advances in inflammaging in liver disease.
Yanping XU ; Luyi CHEN ; Weili LIU ; Liying CHEN
Journal of Zhejiang University. Medical sciences 2025;54(1):90-98
Inflammaging is a process of cellular dysfunction associated with chronic inflammation, which plays a significant role in the onset and progression of liver diseases. Research on its mechanisms has become a hotspot. In viral hepatitis, inflammaging primarily involve oxidative stress, cell apoptosis and necrosis, as well as gut microbiota dysbiosis. In non-alcoholic fatty liver disease, inflammaging is more complex, involving insulin resistance, fat deposition, lipid metabolism disorders, gut microbiota dysbiosis, and abnormalities in NAD+ metabolism. In liver tumors, inflammaging is characterized by weakening of tumor suppressive mechanisms, remodeling of the liver microenvironment, metabolic reprogramming, and enhanced immune evasion. Therapeutic strategies targeting inflammaging have been developing recently, and antioxidant therapy, metabolic disorder improvement, and immunotherapy are emerging as important interventions for liver diseases. This review focuses on the mechanisms of inflammaging in liver diseases, aiming to provide novel insights for the prevention and treatment of liver diseases.
Humans
;
Liver Diseases/pathology*
;
Inflammation
;
Oxidative Stress
;
Non-alcoholic Fatty Liver Disease
;
Liver Neoplasms
;
Gastrointestinal Microbiome
2.Pseudolaric Acid B Alleviates Non-alcoholic Fatty Liver Disease by Targeting PPARα to Regulate Lipid Metabolism and Promote Mitochondrial Biogenesis.
Shu-Yan LIU ; Xiao-Wei ZHANG ; Gai GAO ; Chang-Xin LIU ; Hui CHEN ; Zhong-Xue FU ; Jiang-Yan XU ; Zhen-Zhen WANG ; Zhen-Qiang ZHANG ; Zhi-Shen XIE
Chinese journal of integrative medicine 2025;31(10):877-888
OBJECTIVE:
To investigate the therapeutic potential of pseudolaric acid B (PAB) on non-alcoholic fatty liver disease (NAFLD) and its underlying molecular mechanism in vitro and in vivo.
METHODS:
Eight-week-old male C57BL/6J mice (n=32) were fed either a normal chow diet (NCD) or a high-fat diet (HFD) for 8 weeks. The HFD mice were divided into 3 groups according to a simple random method, including HFD, PAB low-dose [10 mg/(kg·d), PAB-L], and PAB high-dose [20 mg/(kg·d), PAB-H] groups. After 8 weeks of treatment, glucose metabolism and insulin resistance were assessed by oral glucose tolerance test (OGTT) and insulin tolerance test (ITT). Biochemical assays were used to measure the serum and cellular levels of total cholesterol (TC), triglycerides (TG), aspartate aminotransferase (AST), alanine aminotransferase (ALT), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C). White adipose tissue (WAT), brown adipose tissue (BAT) and liver tissue were subjected to hematoxylin and eosin (H&E) staining or Oil Red O staining to observe the alterations in adipose tissue and liver injury. PharmMapper and DisGeNet were used to predict the NAFLD-related PAB targets. Peroxisome proliferator-activated receptor alpha (PPARα) pathway involvement was suggested by Kyoto Encyclopedia of Genes and Genomes (KEGG) and search tool Retrieval of Interacting Genes (STRING) analyses. Luciferase reporter assay, cellular thermal shift assay (CETSA), and drug affinity responsive target stability assay (DARTS) were conducted to confirm direct binding of PAB with PPARα. Molecular dynamics simulations were applied to further validate target engagement. RT-qPCR and Western blot were performed to assess the downstream genes and proteins expression, and validated by PPARα inhibitor MK886.
RESULTS:
PAB significantly reduced serum TC, TG, LDL-C, AST, and ALT levels, and increased HDL-C level in HFD mice (P<0.01). Target prediction analysis indicated a significant correlation between PAB and PPARα pathway. PAB direct target binding with PPARα was confirmed through luciferase reporter assay, CETSA, and DARTS (P<0.05 or P<0.01). The target engagement between PAB and PPARα protein was further confirmed by molecular dynamics simulations and the top 3 amino acid residues, LEU321, MET355, and PHE273 showed the most significant changes in mutational energy. Subsequently, PAB upregulated the genes expressions involved in lipid metabolism and mitochondrial biogenesis downstream of PPARα (P<0.05 or P<0.01). Significantly, the PPARα inhibitor MK886 effectively reversed the lipid-lowering and PPARα activation properties of PAB (P<0.05 or P<0.01).
CONCLUSION
PAB mitigates lipid accumulation, ameliorates liver damage, and improves mitochondrial biogenesis by binding with PPARα, thus presenting a potential candidate for pharmaceutical development in the treatment of NAFLD.
Animals
;
PPAR alpha/metabolism*
;
Non-alcoholic Fatty Liver Disease/pathology*
;
Male
;
Mice, Inbred C57BL
;
Lipid Metabolism/drug effects*
;
Diterpenes/therapeutic use*
;
Organelle Biogenesis
;
Diet, High-Fat
;
Humans
;
Mice
;
Liver/metabolism*
;
Insulin Resistance
;
Mitochondria/metabolism*
;
Molecular Docking Simulation
3.NLRP3 signaling pathway promotes hepatocyte pyroptosis in mice with nonalcoholic steatohepatitis in hypoxic environment.
Shanyu LUO ; Qiang ZHU ; Yufei YAN ; Zonghong JI ; Huajie ZOU ; Ruixia ZHANG ; Yinggui BA
Journal of Southern Medical University 2025;45(9):2026-2033
OBJECTIVES:
To investigate the regulatory role of the NLRP3 signaling pathway in hepatocyte pyroptosis in nonalcoholic steatohepatitis (NASH) under hypoxia.
METHODS:
Twenty-four male C57BL/6 mice were randomized equally into hypoxic control (A), hypoxic NASH model (B), hypoxic NASH+NLRP3 inhibitor (C), and hypoxic NASH+caspase-1 inhibitor (D) groups. In groups B-D, the mice were fed a methionine choline-deficient (MCD) diet under hypoxic conditions (to simulate a 5000 m altitude) for 6 weeks; the mice in groups C and D received intraperitoneal injections of the respective inhibitors every other day.
RESULTS:
Compared with those in group A, the mice in group B showed significantly elevated serum levels of FBG, TC, TG, ALT and AST, increased liver lipid content, inflammatory cell infiltration and collagen fiber deposition, and enhanced hepatic expressions of NLRP3, caspase-1, IL-1β and GSDMD proteins, with obvious swelling, cristae breakage, vacuolization, and outer membrane disruption of the mitochondria, ribosome loss in the cytoplasm, destruction of the nuclear membrane, and pathological changes of the rough endoplasmic reticulum. Treatment with NLRP3 inhibitor and caspase-1 inhibitor both significantly lowered serum levels of TC, TG, ALT and AST (but without significantly affecting FBG) in the mouse models, and reduced liver lipid content, inflammatory cell infiltration, collagen deposition, and expression levels of NLRP3, caspase-1, GSDMD and IL-1β. The treatments also significantly improved pathological changes in the mitochondria, ribosomes and endoplasmic reticulum in liver tissues of the mice.
CONCLUSIONS
NLRP3 signaling pathway plays a key role in promoting hepatocyte pyroptosis in NASH mice under hypoxic condition, and inhibiting this pathway can effectively reduce liver inflammation, suggesting its potential as a therapeutic target for NASH treatment.
Animals
;
Non-alcoholic Fatty Liver Disease/metabolism*
;
NLR Family, Pyrin Domain-Containing 3 Protein/metabolism*
;
Pyroptosis
;
Mice, Inbred C57BL
;
Male
;
Hepatocytes/pathology*
;
Signal Transduction
;
Mice
;
Hypoxia/metabolism*
;
Caspase 1/metabolism*
;
Interleukin-1beta/metabolism*
;
Liver/metabolism*
4.A Novel Signature Combing Cuproptosis- and Ferroptosis-Related Genes in Nonalcoholic Fatty Liver Disease.
Rou-Rou FANG ; Qi-Fan YANG ; Jing ZHAO ; Shou-Zhu XU
Chinese Medical Sciences Journal 2024;39(4):261-272
OBJECTIVES:
To identify cuproptosis- and ferroptosis-related genes involved in nonalcoholic fatty liver disease and to determine the diagnostic value of hub genes.
METHODS:
The gene expression dataset GSE89632 was retrieved from the Gene Expression Omnibus database to identify differentially expressed genes (DEGs) between the non-alcoholic steatohepatitis (NASH) group and the healthy group using the 'limma' package in R software and weighted gene co-expression network analysis. Gene ontology, kyoto encyclopedia of genes and genomes pathway, and single-sample gene set enrichment analyses were performed to identify functional enrichment of DEGs. Ferroptosis- and cuproptosis-related genes were obtained from the FerrDb V2 database and available literatures, respectively. A combined signature for cuproptosis- and ferroptosis-related genes, called CRF, was constructed using the STRING database. Hub genes were identified by overlapping DEGs, WGCNA-derived key genes, and combined signature CRF genes, and validated using the GSE109836 and GSE227714 datasets and real-time quantitative polymerase chain reaction. A nomogram of NASH diagnostic model was established utilizing the 'rms' package in R software based on the hub genes, and the diagnostic value of hub genes was assessed using receiver operating characteristic curve analysis. In addition, immune cell infiltration in NASH versus healthy controls was examined using the CIBERSORT algorithm. The relationships among various infiltrated immune cells were explored with Spearman's correlation analysis.
RESULTS:
Analysis of GSE89632 identified 236 DEGs between the NASH group and the healthy group. WGCNA highlighted 8 significant modules and 11,095 pivotal genes, of which 330 genes constituted CRF. Intersection analysis identified IL6, IL1B, JUN, NR4A1, and PTGS2 as hub genes. The hub genes were all downregulated in the NASH group, and this result was further verified by the NASH validation dataset and real-time quantitative polymerase chain reaction. Receiver operating characteristic curve analysis confirmed the diagnostic efficacy of these hub genes with areas under the curve of 0.985, 0.941, 1.000, 0.967, and 0.985, respectively. Immune infiltration assessment revealed that gamma delta T cells, M1 macrophages, M2 macrophages, and resting mast cells were predominantly implicated.
CONCLUSIONS
Our investigation underscores the significant association of cuproptosis- and ferroptosis-related genes, specifically IL6, IL1B, JUN, NR4A1, and PTGS2, with NASH. These findings offer novel insights into the pathogenesis of NASH, potentially guiding future diagnostic and therapeutic strategies.
Non-alcoholic Fatty Liver Disease/pathology*
;
Humans
;
Ferroptosis/genetics*
;
Copper/metabolism*
;
Gene Ontology
;
Gene Expression Profiling
5.Role of AMPKα2 in regulating the IRE1α-JNK pathway in metabolic dysfunction-associated fatty liver disease.
Shujie ZHAO ; Weilun FANG ; Yu WEI ; Jiahui MENG ; Qiao JIN ; Weijin FANG
Journal of Central South University(Medical Sciences) 2024;49(12):1891-1901
OBJECTIVES:
Over 25% of the global population is affected by metabolic dysfunction-associated fatty liver disease (MAFLD), yet its pathogenesis remains unclear. Endoplasmic reticulum stress (ERS) may be involved in the onset and progression of MAFLD. Adenosine 5'-monophosphate-activated protein kinase α2 (AMPKα2), a key regulator of hepatic energy metabolism, may influence MAFLD development via ERS modulation. This study aims to investigate the role of AMPKα2 in a high-fat diet-induced MAFLD mouse model and its regulatory effect on the inositol-requiring enzyme 1 alpha (IRE1α)-c-Jun N-terminal kinase (JNK) signaling pathway.
METHODS:
Liver-specific AMPKα2 knockout mice on a C57BL/6 background were generated and subjected to MAFLD induction. Mice were divided into four groups: wild-type control (WT+Chow, basic diet for 12 weeks), wild-type high-fat diet (WT+HFD, high-fat diet for 12 weeks), AMPKα2 knockout control (AMPKα2 KO+Chow), and AMPKα2 knockout high-fat diet (AMPKα2 KO+HFD). Blood glucose, lipid levels, and liver function were assessed post-treatment. Liver histology was analyzed using Oil Red O, hematoxylin-eosin, Masson, and Sirius Red staining. Western blotting was used to evaluate the expression of AMPKα2, ERS markers, autophagy, apoptosis, and ferroptosis-related proteins.
RESULTS:
Compared with the WT+Chow group, the WT+HFD group showed significantly elevated blood glucose, alanine aminotransferase (ALT), and aspartate aminotransferase (AST) levels (all P<0.01); histological analyses revealed hepatic steatosis, vacuolization, and fibrosis, with a significantly increased non-alcoholic steatohepatitis activity score (NAS) (P<0.001). Phosphorylated IRE1α and the autophagy marker microtubule-associated protein light chain (LC) 3II/LC3I were markedly upregulated, while apoptotic proteins (Cleaved-Caspase 3, BAX, Bcl-2) and ferroptosis markers (SLC7A11, GPX4) showed no significant change (P>0.05). In the AMPKα2 KO+HFD group, blood glucose, ALT, and AST levels were significantly reduced compared to the WT+HFD group. Histological improvements were observed with reduced vacuolization and lipid accumulation. Expression of p-IRE1α, JNK, and LC3II/LC3I was significantly decreased (P<0.05).
CONCLUSIONS
Hepatic AMPKα2 knockout alleviates high-fat induced MAFLD, potentially by inhibiting the IRE1α-JNK pathway and reducing autophagy.
Animals
;
AMP-Activated Protein Kinases/physiology*
;
Protein Serine-Threonine Kinases/metabolism*
;
Mice, Knockout
;
Diet, High-Fat/adverse effects*
;
Mice, Inbred C57BL
;
Mice
;
Endoplasmic Reticulum Stress
;
Endoribonucleases/metabolism*
;
Male
;
Liver/pathology*
;
Non-alcoholic Fatty Liver Disease/metabolism*
;
MAP Kinase Signaling System/physiology*
;
Fatty Liver/metabolism*
;
Signal Transduction
6.Research progress of celastrol on the prevention and treatment of metabolic associated fatty liver disease.
Yun-Chao LIU ; Ying ZHANG ; Shu-Cun QIN ; Jun-Li XUE
Acta Physiologica Sinica 2023;75(5):682-690
Metabolic associated fatty liver disease (MAFLD) is a liver disease with hepatocyte steatosis caused by metabolic disorders, which is closely related to obesity, diabetes, metabolic dysfunction, and other factors. Its pathological process changes from simple steatosis, liver inflammation to non-alcoholic steatohepatitis (NASH), and then leads to liver fibrosis, cirrhosis, and liver cancer. At present, no specific therapeutics are available for treatment of MAFLD targeting its etiology. Celastrol is the main active component of the traditional Chinese medicine Celastrus orbiculatus Thunb. In recent years, it has been found that celastrol shows important medicinal value in regulating lipid metabolism, reducing fat and weight, and protecting liver, and then ameliorates MAFLD. This article reviews the related research progress of celastrol in the prevention and treatment of MAFLD, so as to provide a reference for the comprehensive development and utilization of celastrol.
Humans
;
Non-alcoholic Fatty Liver Disease/metabolism*
;
Liver/pathology*
;
Pentacyclic Triterpenes/metabolism*
;
Obesity
7.The role of apolipoprotein C3 in the regulation of nonalcoholic fatty liver disease, glucose and lipid metabolism, and islet β cell function.
Shan YAN ; Zhi-Yong DING ; Yuan GAO ; Wang-Jia MAO ; Xiao-Yun CHENG
Acta Physiologica Sinica 2023;75(6):767-778
As a member of the apolipoprotein C (ApoC) family with a relatively high content, ApoC3 plays a major role in the regulation of triglyceride metabolism, and plays an important role in the occurrence and development of cardiovascular diseases, glucose and lipid metabolism disorders. Nonalcoholic fatty liver disease (NAFLD) refers to the accumulation of a large amount of fat in the liver in the absence of a history of chronic alcohol consumption or other damage to the liver. A large number of previous studies have shown that there is a correlation between the gene polymorphism and high expression of ApoC3 and NAFLD. In the context of hypertriglyceridemia (HTG), this article reviews the relationship between ApoC3 and NAFLD, glucose and lipid metabolism, and islet β cell function, showing that ApoC3 can not only inhibit lipoprotein lipase (LPL) and hepatic lipase (HL) activity, delay the decomposition of triglyceride in plasma to maintain the body's energy metabolism during fasting, but also be significantly increased under insulin resistance, prompting the liver to secrete a large amount of very low-density lipoprotein (VLDL) to induce HTG. Therefore, targeting and inhibiting ApoC3 might become a new approach to treat HTG. Increasing evidence suggests that ApoC3 does not appear to be an independent "contributor" to NAFLD. Similarly, our previous studies have shown that ApoC3 is not an independent factor triggering islet β cell dysfunction in ApoC3 transgenic mice, but in a state of excess nutrition, HTG triggered by ApoC3 high expression may exacerbate the effects of hyperglycemia and insulin resistance on islet β cell function, and the underlying mechanism remains to be further discussed.
Apolipoprotein C-III/genetics*
;
Non-alcoholic Fatty Liver Disease/pathology*
;
Glucose/metabolism*
;
Lipid Metabolism
;
Humans
;
Animals
;
Hypertriglyceridemia/metabolism*
;
Islets of Langerhans/metabolism*
8.Liver macrophages show an immunotolerance phenotype in nonalcoholic fatty liver combined with Porphyromonas gingivalis-lipopolysaccharide infection.
Lijia GUO ; Yitong LIU ; Yingyi CHEN ; Junji XU ; Yi LIU
West China Journal of Stomatology 2023;41(4):385-394
OBJECTIVES:
This study aimed to explore the functions and potential regulatory targets of local macrophages in nonalcoholic fatty liver combined with Porphyromonas gingivalis (P. gingivalis)infection.
METHODS:
Single-cell RNA sequencing was used to analyze the phenotypes and functional changes in various cells in the liver tissue of nonalcoholic steatohepatitis (NASH) mice fed with P. gingivalis. Real-time polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay, and immunofluorescence staining were applied to observe the inflammation and expression levels of macrophage antigen presenting functional markers in the NASH liver. Oil red staining was performed to observe the accumulation of local adipose tissue in the NASH liver. Results were verified through RT-PCRand RNA sequencing using P. gingivalis-lipopolysaccharide treated mouse peritoneal macrophages.
RESULTS:
In comparison with healthy livers with Kupffer cells, the NASH liver combined with P. gingivalis infection-related macrophages showed significant heterogeneity. C1qb, C1qc, Mafb, Apoe, and Cd14 were highly expressed, but Cd209a, H2-Aa, H2-Ab1, and H2-DMb1, which are related to the antigen presentation function, were weakly expressed. Further in vivo and in vitro investigations indicated that the activation and infiltration of these macrophages may be due to local P. gingivalis-lipopolysaccharide accumulation.
CONCLUSIONS
P. gingivalis-lipopolysaccharide induces a local macrophage immunotolerance phenotype in nonalcoholic fatty liver, which may be the key mechanism of periodontitis pathogen infection that promotes NASH inflammation and pathogenesis. This study further clarifies the dysfunction and regulatory mechanisms of macrophages in the pathogenesis of P. gingivalis-infected NASH, thereby providing potential therapeutic targets for its clinical treatment.
Mice
;
Animals
;
Non-alcoholic Fatty Liver Disease/pathology*
;
Kupffer Cells/pathology*
;
Porphyromonas gingivalis
;
Lipopolysaccharides/metabolism*
;
Inflammation/pathology*
;
Macrophages/metabolism*
;
Mice, Inbred C57BL
9.Analysis of clinicopathological characteristics of non-alcoholic fatty liver disease.
Jiao LI ; Ye Fan YANG ; Miao Miao SUN ; Ying Yun GONG ; Hong Wen ZHOU ; Zhi Hong ZHANG
Chinese Journal of Pathology 2023;52(5):486-491
Objective: To investigate the clinical and pathologic characteristics of obese adults with nonalcoholic fatty liver disease (NAFLD) and to aid the diagnosis of nonalcoholic steatohepatitis (NASH). Methods: A total of 262 patients eligible for inclusion who received volume reduction metabolism surgery and liver biopsy in the First Affiliated Hospital of Nanjing Medical University from June 2018 to September 2019 were selected. HE staining, reticular fiber staining and Masson staining were performed. Statistical analysis was performed using SPSS 20.0. Results: The patients ranged in age from 18 to 66 years. Among the 262 cases, 65 cases (65/262, 24.8%) were male and 197 cases (197/262, 75.2%) were female. Sixty-one cases (61/262, 23.3%) were non-NAFLD, 201 cases (201/262, 76.7%) were NAFLD including 27 cases (27/201, 13.4%) of nonalcoholic fatty live (NAFL) and 174 cases (174/201, 86.6%) of NASH. The main lesions of NAFLD were in hepatic acinus zone 3. There were significant differences in age, alanine aminotransferase (ALT), glutamic oxaloacetic transaminase (AST), body mass index (BMI), fasting blood-glucose (FPG) and apolipoprotein A (APOA) levels among the non-NAFLD group, NAFL group and NASH group (P<0.05). Patients with BMI≥35 m/kg2 combined with type 2 diabetes had a higher prevalence of NASH. Multiple logistic regression showed that ALT and APOA were independent predictors of NASH (P<0.001, OR=1.05, 95%CI: 1.020-1.082; P=0.027, OR=0.916, 95%CI: 0.878-0.941). Total cholesterol (CHO) and high-density lipoprotein (HDL) were independent predictors of lobular inflammation (P=0.043, 95%CI: 0.010-0.634; P=0.024, 95%CI:-3.068--0.216). AST and HDL were independent predictors of fibrosis stage (P=0.029, 95%CI: 0.001-0.021; P<0.001, 95%CI:-2.670--0.645). Conclusions: Biochemical indicators of NAFLD are closely related to its pathology. The histological lesions of NAFLD are mainly present in hepatic acinar area 3. The diagnosis of NASH is supported by extensive steatosis and high levels of CHO, ALT, AST and BMI, low levels of HDL and ApoA in biochemical markers, but pathological examination is still the gold standard for it.
Adult
;
Humans
;
Male
;
Female
;
Adolescent
;
Young Adult
;
Middle Aged
;
Aged
;
Non-alcoholic Fatty Liver Disease/pathology*
;
Diabetes Mellitus, Type 2/pathology*
;
Liver/pathology*
;
Obesity/pathology*
;
Apolipoproteins A
10.Analysis of clinical characteristics and risk factors of hepatic fibrosis in children with chronic hepatitis B combined with metabolic-related fatty liver disease.
Wwei LI ; Li Na JIANG ; Bo Kang ZHAO ; Hong Yang LIU ; Jing Min ZHAO
Chinese Journal of Hepatology 2023;31(6):601-607
Objective: To compare the clinical and pathological features of children with chronic viral hepatitis B combined with metabolic-associated fatty liver disease (CHB-MAFLD) and chronic viral hepatitis B alone (CHB alone), and to further explore the effect of MAFLD on the progression of hepatic fibrosis in CHB. Methods: 701 initially treated CHB children confirmed by liver biopsy admitted to the Fifth Medical Center of the PLA General Hospital from January 2010 to December 2021 were collected continuously. They were divided into CHB-MAFLD and CHB-alone groups according to whether they were combined with MAFLD. A retrospective case-control study was conducted. CHB-MAFLD was used as the case group, and 1:2 propensity score matching was performed with the CHB alone group according to age and gender, including 56 cases in the CHB-MAFLD group and 112 cases in the CHB alone group. The body mass index (BMI), metabolic complications, laboratory indicators, and pathological characteristics of liver tissue were compared between the two groups. The related factors affecting liver disease progression in CHB were analyzed by a binary logistic regression model. The measurement data between groups were compared using the t-test and rank sum test. The χ (2) test was used for the comparison of categorical data between groups. Results: Alanine aminotransferase (ALT, P = 0.032) and aspartate aminotransferase (AST, P = 0.003) levels were lower in the CHB-MAFLD group than those in the CHB alone group, while BMI (P < 0.001), triglyceride (TG, P < 0.001), total cholesterol (P = 0.016) and the incidence of metabolic syndrome (P < 0.001) were higher in the CHB alone group. There were no statistically significant differences in HBsAg quantification or HBV DNA load between the two groups (P > 0.05). Histologically, the proportion of significant liver fibrosis (S2-S4) was higher in the CHB-MAFLD group than that in the CHB alone group (67.9% vs. 49.1%, χ (2) = 5.311, P = 0.021). Multivariate regression results showed that BMI (OR = 1.258, 95% CI: 1.145 ~ 1.381, P = 0.001) and TG (OR = 12.334, 95% CI: 3.973 ~ 38.286, P < 0.001) were the risk factors for hepatic steatosis occurrence in children with CHB. MAFLD (OR = 4.104, 95% CI: 1.703 ~ 9.889, P = 0.002), liver inflammation (OR = 3.557, 95% CI: 1.553 ~ 8.144, P = 0.003), and γ-glutamyl transferase (OR = 1.019, 95% CI: 1.001 to 1.038, P = 0.038) were independent risk factors for significant hepatic fibrosis in children with CH. Conclusion: MAFLD occurrence is related to metabolic factors in children with CHB. Additionally, the combination of MAFLD may promote liver fibrosis progression in CHB patients.
Humans
;
Child
;
Hepatitis B, Chronic/pathology*
;
Retrospective Studies
;
Case-Control Studies
;
Hepatitis B virus/genetics*
;
Liver Cirrhosis/pathology*
;
Non-alcoholic Fatty Liver Disease/complications*
;
Risk Factors

Result Analysis
Print
Save
E-mail