Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.283
Filtrer
1.
Lipids Health Dis ; 23(1): 249, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39148061

RÉSUMÉ

BACKGROUND: Existing studies have presented limited and disparate findings on the nexus between immune cells, plasma metabolites, and metabolic dysfunction-associated steatotic liver disease (MASLD). The aim of this study was to investigate the causal relationship between immune cells and MASLD. Additionally, we aimed to identify and quantify the potential mediating role of metabolites. METHODS: A Mendelian randomization (MR) analysis was conducted using two samples of pooled data from genome-wide association studies on MASLD that included 2568 patients and 409,613 control individuals. Additionally, a mediated MR study was employed to quantify the metabolite-mediated immune cell effects on MASLD. RESULTS: In this study, eight immunophenotypes were linked to the risk of MASLD, and thirty-five metabolites/metabolite ratios were linked to the occurrence of MASLD. Furthermore, a total of six combinations of immunophenotypic and metabolic factors demonstrated effects on the occurrence of MASLD, although the mediating effects of metabolites were not significant. CONCLUSION: Our study demonstrated that certain immunophenotypes and metabolite/metabolite ratios have independent causal relationships with MASLD. Furthermore, we identified specific metabolites/metabolite ratios that are associated with an increased risk of MASLD. However, their mediating role in the causal association between immunophenotypes and MASLD was not significant. It is important to consider immune and metabolic disorders among patients with MASLD in clinical practice.


Sujet(s)
Étude d'association pangénomique , Analyse de randomisation mendélienne , Humains , Polymorphisme de nucléotide simple , Stéatose hépatique/génétique , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Stéatose hépatique/immunologie , Immunophénotypage , Mâle
2.
Article de Anglais | MEDLINE | ID: mdl-39142818

RÉSUMÉ

Genetic susceptibility to metabolic associated fatty liver disease (MAFLD) is complex and poorly characterized. Accurate characterization of the genetic background of hepatic fat content would provide insights into disease etiology and causality of risk factors. We performed genome-wide association study (GWAS) on two noninvasive definitions of hepatic fat content: magnetic resonance imaging proton density fat fraction (MRI-PDFF) in 16,050 participants and fatty liver index (FLI) in 388,701 participants from the United Kingdom (UK) Biobank (UKBB). Heritability, genetic overlap, and similarity between hepatic fat content phenotypes were analyzed, and replicated in 10,398 participants from the University Medical Center Groningen (UMCG) Genetics Lifelines Initiative (UGLI). Meta-analysis of GWASs of MRI-PDFF in UKBB revealed five statistically significant loci, including two novel genomic loci harboring CREB3L1 (rs72910057-T, P = 5.40E-09) and GCM1 (rs1491489378-T, P = 3.16E-09), respectively, as well as three previously reported loci: PNPLA3, TM6SF2, and APOE. GWAS of FLI in UKBB identified 196 genome-wide significant loci, of which 49 were replicated in UGLI, with top signals in ZPR1 (P = 3.35E-13) and FTO (P = 2.11E-09). Statistically significant genetic correlation (rg) between MRI-PDFF (UKBB) and FLI (UGLI) GWAS results was found (rg = 0.5276, P = 1.45E-03). Novel MRI-PDFF genetic signals (CREB3L1 and GCM1) were replicated in the FLI GWAS. We identified two novel genes for MRI-PDFF and 49 replicable loci for FLI. Despite a difference in hepatic fat content assessment between MRI-PDFF and FLI, a substantial similar genetic architecture was found. FLI is identified as an easy and reliable approach to study hepatic fat content at the population level.


Sujet(s)
Prédisposition génétique à une maladie , Étude d'association pangénomique , Foie , Humains , Femelle , Mâle , Facteurs de risque , Prédisposition génétique à une maladie/génétique , Foie/imagerie diagnostique , Foie/métabolisme , Foie/anatomopathologie , Adulte d'âge moyen , Polymorphisme de nucléotide simple/génétique , Imagerie par résonance magnétique , Stéatose hépatique non alcoolique/génétique , Stéatose hépatique non alcoolique/imagerie diagnostique , Adulte , Sujet âgé , Stéatose hépatique/génétique , Stéatose hépatique/imagerie diagnostique
3.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39125617

RÉSUMÉ

Progression of metabolic dysfunction-associated steatites liver disease (MASLD) to steatohepatitis (MASH) is driven by stress-inducing lipids that promote liver inflammation and fibrosis, and MASH can lead to cirrhosis and hepatocellular carcinoma. Previously, we showed coordinated defenses regulated by transcription factors, nuclear factor erythroid 2-related factor-1 (Nrf1) and -2 (Nrf2), protect against hepatic lipid stress. Here, we investigated protective effects of hepatocyte Nrf1 and Nrf2 against MASH-linked liver fibrosis and tumorigenesis. Male and female mice with flox alleles for genes encoding Nrf1 (Nfe2l1), Nrf2 (Nfe2l2), or both were fed a MASH-inducing diet enriched with high fat, fructose, and cholesterol (HFFC) or a control diet for 24-52 weeks. During this period, hepatocyte Nrf1, Nrf2, or combined deficiency for ~7 days, ~7 weeks, and ~35 weeks was induced by administering mice hepatocyte-targeting adeno-associated virus (AAV) expressing Cre recombinase. The effects on MASH, markers of liver fibrosis and proliferation, and liver tumorigenesis were compared to control mice receiving AAV-expressing green fluorescent protein. Also, to assess the impact of Nrf1 and Nrf2 induction on liver fibrosis, HFFC diet-fed C57bl/6J mice received weekly injections of carbon tetrachloride, and from week 16 to 24, mice were treated with the Nrf2-activating drug bardoxolone, hepatocyte overexpression of human NRF1 (hNRF1), or both, and these groups were compared to control. Compared to the control diet, 24-week feeding with the HFFC diet increased bodyweight as well as liver weight, steatosis, and inflammation. It also increased hepatocyte proliferation and a marker of liver damage, p62. Hepatocyte Nrf1 and combined deficiency increased liver steatosis in control diet-fed but not HFFC diet-fed mice, and increased liver inflammation under both diet conditions. Hepatocyte Nrf1 deficiency also increased hepatocyte proliferation, whereas combined deficiency did not, and this also occurred for p62 level in control diet-fed conditions. In 52-week HFFC diet-fed mice, 35 weeks of hepatocyte Nrf1 deficiency, but not combined deficiency, resulted in more liver tumors in male mice, but not in female mice. In contrast, hepatocyte Nrf2 deficiency had no effect on any of these parameters. However, in the 15-week CCL4-exposed and 24-week HFFC diet-fed mice, Nrf2 induction with bardoxolone reduced liver steatosis, inflammation, fibrosis, and proliferation. Induction of hepatic Nrf1 activity with hNRF1 enhanced the effect of bardoxolone on steatosis and may have stimulated liver progenitor cells. Physiologic Nrf1 delays MASLD progression, Nrf2 induction alleviates MASH, and combined enhancement synergistically protects against steatosis and may facilitate liver repair.


Sujet(s)
Hépatocytes , Facteur-2 apparenté à NF-E2 , Animaux , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Souris , Hépatocytes/métabolisme , Mâle , Femelle , Évolution de la maladie , Souris de lignée C57BL , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Stéatose hépatique/génétique , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Cirrhose du foie/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Facteur-1 apparenté à NF-E2/métabolisme , Facteur-1 apparenté à NF-E2/génétique , Facteur nucléaire-1 respiratoire/métabolisme , Facteur nucléaire-1 respiratoire/génétique , Alimentation riche en graisse/effets indésirables , Foie/métabolisme , Foie/anatomopathologie , Humains
4.
Elife ; 122024 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-39037913

RÉSUMÉ

Background: The development of obesity-associated comorbidities such as type 2 diabetes (T2D) and hepatic steatosis has been linked to selected microRNAs in individual studies; however, an unbiased genome-wide approach to map T2D induced changes in the miRNAs landscape in human liver samples, and a subsequent robust identification and validation of target genes are still missing. Methods: Liver biopsies from age- and gender-matched obese individuals with (n=20) or without (n=20) T2D were used for microRNA microarray analysis. The candidate microRNA and target genes were validated in 85 human liver samples, and subsequently mechanistically characterized in hepatic cells as well as by dietary interventions and hepatic overexpression in mice. Results: Here, we present the human hepatic microRNA transcriptome of type 2 diabetes in liver biopsies and use a novel seed prediction tool to robustly identify microRNA target genes, which were then validated in a unique cohort of 85 human livers. Subsequent mouse studies identified a distinct signature of T2D-associated miRNAs, partly conserved in both species. Of those, human-murine miR-182-5 p was the most associated with whole-body glucose homeostasis and hepatic lipid metabolism. Its target gene LRP6 was consistently lower expressed in livers of obese T2D humans and mice as well as under conditions of miR-182-5 p overexpression. Weight loss in obese mice decreased hepatic miR-182-5 p and restored Lrp6 expression and other miR-182-5 p target genes. Hepatic overexpression of miR-182-5 p in mice rapidly decreased LRP6 protein levels and increased liver triglycerides and fasting insulin under obesogenic conditions after only seven days. Conclusions: By mapping the hepatic miRNA-transcriptome of type 2 diabetic obese subjects, validating conserved miRNAs in diet-induced mice, and establishing a novel miRNA prediction tool, we provide a robust and unique resource that will pave the way for future studies in the field. As proof of concept, we revealed that the repression of LRP6 by miR-182-5 p, which promotes lipogenesis and impairs glucose homeostasis, provides a novel mechanistic link between T2D and non-alcoholic fatty liver disease, and demonstrate in vivo that miR-182-5 p can serve as a future drug target for the treatment of obesity-driven hepatic steatosis. Funding: This work was supported by research funding from the Deutsche Forschungsgemeinschaft (KI 1887/2-1, KI 1887/2-2, KI 1887/3-1 and CRC-TR296), the European Research Council (ERC, CoG Yoyo LepReSens no. 101002247; PTP), the Helmholtz Association (Initiative and Networking Fund International Helmholtz Research School for Diabetes; MB) and the German Center for Diabetes Research (DZD Next Grant 82DZD09D1G).


Sujet(s)
Diabète de type 2 , Foie , microARN , Obésité , Transcriptome , microARN/métabolisme , microARN/génétique , Diabète de type 2/génétique , Diabète de type 2/métabolisme , Animaux , Humains , Obésité/génétique , Obésité/métabolisme , Foie/métabolisme , Souris , Mâle , Stéatose hépatique/génétique , Stéatose hépatique/métabolisme , Femelle , Souris de lignée C57BL , Adulte d'âge moyen , Analyse de profil d'expression de gènes
5.
Zhongguo Zhong Yao Za Zhi ; 49(13): 3600-3607, 2024 Jul.
Article de Chinois | MEDLINE | ID: mdl-39041132

RÉSUMÉ

Based on the Toll-like receptor 4(TLR4)/myeloid differentiation factor 88(MyD88)/nuclear factor kappaB(NF-κB) signaling pathway, this study observed the regulatory effect of ginsenoside Rb_1(Rb_1) on liver lipid metabolism in db/db obese mice and explored its potential mechanism. Thirty 6-week-old male db/db mice were randomly divided into a model group, a metformin group, and Rb_1 groups with low, medium, and high doses, with six mice in each group. Additionally, six age-matched male db/m mice were assigned to the normal group. The intervention lasted for five weeks. Body weight, fasting blood glucose, and food intake were mea-sured weekly. At the end of the experiment, serum lipid levels and liver function were detected. Hematoxylin-eosin(HE) staining and oil red O staining were performed to observe pathological changes in liver tissue. Real-time quantitative PCR and immunohistochemistry on paraffin sections were used to detect the mRNA and protein expression of TLR4, MyD88, and NF-κB p65. RESULTS:: showed that compared with the normal group, the model group exhibited significant increases in body weight, liver weight, liver index, epididymal fat mass, epididymal fat index, total cholesterol, low-density lipoprotein cholesterol, liver function parameters, and fasting blood glucose levels. Liver lipid accumulation significantly increased, along with elevated mRNA and protein expression of TLR4, MyD88, and NF-κB p65 in the liver. After Rb_1 treatment, the above-mentioned parameters in the intervention groups showed significant reversals. In conclusion, Rb_1 can improve obesity and obesity-related hepatic steatosis in mice while regulating abnormal lipid and glucose meta-bolism. Mechanistically, Rb_1 may improve liver steatosis in db/db obese mice by modulating the TLR4/MyD88/NF-κB signaling pathway.


Sujet(s)
Stéatose hépatique , Ginsénosides , Facteur de différenciation myéloïde-88 , Facteur de transcription NF-kappa B , Transduction du signal , Récepteur de type Toll-4 , Animaux , Ginsénosides/pharmacologie , Ginsénosides/administration et posologie , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/métabolisme , Facteur de différenciation myéloïde-88/génétique , Facteur de différenciation myéloïde-88/métabolisme , Souris , Mâle , Facteur de transcription NF-kappa B/génétique , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Stéatose hépatique/traitement médicamenteux , Stéatose hépatique/métabolisme , Stéatose hépatique/génétique , Obésité/traitement médicamenteux , Obésité/métabolisme , Obésité/génétique , Souris obèse , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Humains , Médicaments issus de plantes chinoises/administration et posologie , Médicaments issus de plantes chinoises/pharmacologie
6.
Hepatol Commun ; 8(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38967582

RÉSUMÉ

BACKGROUND: Fibrosis-4 (FIB4) is a recommended noninvasive test to assess hepatic fibrosis among patients with metabolic dysfunction-associated steatotic liver disease (MASLD). Here, we used FIB4 trajectory over time (ie, "slope" of FIB4) as a surrogate marker of liver fibrosis progression and examined if FIB4 slope is associated with clinical and genetic factors among individuals with clinically defined MASLD within the Million Veteran Program Cohort. METHODS: In this retrospective cohort study, FIB4 slopes were estimated through linear regression for participants with clinically defined MASLD and FIB4 <2.67 at baseline. FIB4 slope was correlated with demographic parameters and clinical outcomes using logistic regression and Cox proportional hazard models. FIB4 slope as a quantitative phenotype was used in a genome-wide association analysis in ancestry-specific analysis and multiancestry meta-analysis using METAL. RESULTS: FIB4 slopes, generated from 98,361 subjects with MASLD (16,045 African, 74,320 European, and 7996 Hispanic), showed significant associations with sex, ancestry, and cardiometabolic risk factors (p < 0.05). FIB4 slopes also correlated strongly with hepatic outcomes and were independently associated with time to cirrhosis. Five genetic loci showed genome-wide significant associations (p < 5 × 10-8) with FIB4 slope among European ancestry subjects, including 2 known (PNPLA3 and TM6SF2) and 3 novel loci (TERT 5.1 × 10-11; LINC01088, 3.9 × 10-8; and MRC1, 2.9 × 10-9). CONCLUSIONS: Linear trajectories of FIB4 correlated significantly with time to progression to cirrhosis, with liver-related outcomes among individuals with MASLD and with known and novel genetic loci. FIB4 slope may be useful as a surrogate measure of fibrosis progression.


Sujet(s)
Évolution de la maladie , Étude d'association pangénomique , Cirrhose du foie , Humains , Mâle , Femelle , Cirrhose du foie/génétique , Cirrhose du foie/complications , Adulte d'âge moyen , Études rétrospectives , Facteurs de risque , Sujet âgé , Protéines membranaires/génétique , Stéatose hépatique/génétique , Marqueurs biologiques , Stéatose hépatique non alcoolique/génétique , Stéatose hépatique non alcoolique/complications , Acyltransferases , Calcium-independent phospholipase A2
7.
J Transl Med ; 22(1): 650, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997780

RÉSUMÉ

BACKGROUND: Although the inherited risk factors associated with fatty liver disease are well understood, little is known about the genetic background of metabolic dysfunction-associated steatotic liver disease (MASLD) and its related health impacts. Compared to non-alcoholic fatty liver disease (NAFLD), MASLD presents significantly distinct diagnostic criteria, and epidemiological and clinical features, but the related genetic variants are yet to be investigated. Therefore, we conducted this study to assess the genetic background of MASLD and interactions between MASLD-related genetic variants and metabolism-related outcomes. METHODS: Participants from the UK Biobank were grouped into discovery and replication cohorts for an MASLD genome-wide association study (GWAS), and base and target cohorts for polygenic risk score (PRS) analysis. Autosomal genetic variants associated with NAFLD were compared with the MASLD GWAS results. Kaplan-Meier and Cox regression analyses were used to assess associations between MASLD and metabolism-related outcomes. RESULTS: Sixteen single-nucleotide polymorphisms (SNPs) were identified at genome-wide significance levels for MASLD and duplicated in the replication cohort. Differences were found after comparing these SNPs with the results of NAFLD-related genetic variants. MASLD cases with high PRS had a multivariate-adjusted hazard ratio of 3.15 (95% confidence interval, 2.54-3.90) for severe liver disease (SLD), and 2.81 (2.60-3.03) for type 2 diabetes mellitus. The high PRS amplified the impact of MASLD on SLD and extrahepatic outcomes. CONCLUSIONS: High PRS of MASLD GWAS amplified the impact of MASLD on SLD and metabolism-related outcomes, thereby refining the process of identification of individuals at high risk of MASLD. Supplementation of this process with relevant genetic backgrounds may lead to more effective MASLD prevention and management.


Sujet(s)
Prédisposition génétique à une maladie , Étude d'association pangénomique , Hérédité multifactorielle , Polymorphisme de nucléotide simple , Humains , Polymorphisme de nucléotide simple/génétique , Mâle , Femelle , Hérédité multifactorielle/génétique , Facteurs de risque , Adulte d'âge moyen , Stéatose hépatique/génétique , Stéatose hépatique/complications , Stéatose hépatique non alcoolique/génétique , Maladies métaboliques/génétique , Maladies métaboliques/complications , Études de cohortes , Estimation de Kaplan-Meier , Sujet âgé , Modèles des risques proportionnels ,
8.
BMC Pulm Med ; 24(1): 368, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39080576

RÉSUMÉ

BACKGROUND: Hepatic steatosis and its related complications are risk factors for multiple respiratory diseases; however, the causal relationship between metabolic dysfunction-associated steatotic liver disease (MASLD) and pulmonary function remains controversial. We aimed to identify it using a national cohort and Mendelian randomization (MR). METHODS: We enrolled 30,442 participants from the 2007 to 2012 National Health and Nutrition Examination Survey. Demographics, pulmonary function indices (forced expiratory volume in 1 s [FEV1], forced vital capacity [FVC]), and variables used to calculate the liver fat score (LFS) were collected. A two-sample MR analysis employing the summary data of genome-wide association studies on MASLD and FEV1/FVC, chronic obstructive pulmonary disease (COPD), and asthma from the Finngen Biobank and Medical Research Council Integrative Epidemiology Unit was performed. RESULTS: A total of 3,462 participants, 1,335 of whom had MASLD (LFS > -0.640), were finally included in the study. The FEV1 (3,204.7 vs. 3,262.5 ml, P = 0.061), FVC (4,089.1 vs. 4,143.8 ml, P = 0.146), FEV1/FVC ratio (78.5% vs. 78.8%, P = 0.233), and FEV1/predicted FEV1 ratio (146.5% vs. 141.7%, P = 0.366) were not significantly different between people with MASLD and those without. Additionally, the MR analysis suggested no causal correlation between MASLD and FEV1/FVC (P = 0.817), MASLD and COPD (P = 0.407), and MASLD and asthma (P = 0.808). Reverse MR studies showed no causal relationships yet (all P > 0.05). CONCLUSION: Our study provides convincing evidence that there is no causal association between MASLD and pulmonary function.


Sujet(s)
Asthme , Étude d'association pangénomique , Analyse de randomisation mendélienne , Enquêtes nutritionnelles , Broncho-pneumopathie chronique obstructive , Humains , Mâle , Femelle , Adulte d'âge moyen , Asthme/génétique , Asthme/physiopathologie , Asthme/épidémiologie , Broncho-pneumopathie chronique obstructive/génétique , Broncho-pneumopathie chronique obstructive/physiopathologie , Broncho-pneumopathie chronique obstructive/épidémiologie , Capacité vitale , Volume expiratoire maximal par seconde , Sujet âgé , Adulte , Facteurs de risque , Poumon/physiopathologie , Stéatose hépatique/génétique , Stéatose hépatique/physiopathologie , Tests de la fonction respiratoire
9.
Cardiovasc Diabetol ; 23(1): 228, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951793

RÉSUMÉ

BACKGROUND: Glucokinase (GK) plays a key role in glucose metabolism. In the liver, GK is regulated by GK regulatory protein (GKRP) with nuclear sequestration at low plasma glucose level. Some GK activators (GKAs) disrupt GK-GKRP interaction which increases hepatic cytoplasmic GK level. Excess hepatic GK activity may exceed the capacity of glycogen synthesis with excess triglyceride formation. It remains uncertain whether hypertriglyceridemia associated with some GKAs in previous clinical trials was due to direct GK activation or impaired GK-GKRP interaction. METHODS: Using publicly available genome-wide association study summary statistics, we selected independent genetic variants of GCKR and GCK associated with fasting plasma glucose (FPG) as instrumental variables, to mimic the effects of impaired GK-GKRP interaction and direct GK activation, respectively. We applied two-sample Mendelian Randomization (MR) framework to assess their causal associations with lipid-related traits, risks of metabolic dysfunction-associated steatotic liver disease (MASLD) and cardiovascular diseases. We verified these findings in one-sample MR analysis using individual-level statistics from the Hong Kong Diabetes Register (HKDR). RESULTS: Genetically-proxied impaired GK-GKRP interaction increased plasma triglycerides, low-density lipoprotein cholesterol and apolipoprotein B levels with increased odds ratio (OR) of 14.6 (95% CI 4.57-46.4) per 1 mmol/L lower FPG for MASLD and OR of 2.92 (95% CI 1.78-4.81) for coronary artery disease (CAD). Genetically-proxied GK activation was associated with decreased risk of CAD (OR 0.69, 95% CI 0.54-0.88) and not with dyslipidemia. One-sample MR validation in HKDR showed consistent results. CONCLUSIONS: Impaired GK-GKRP interaction, rather than direct GK activation, may worsen lipid profiles and increase risks of MASLD and CAD. Development of future GKAs should avoid interfering with GK-GKRP interaction.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Glycémie , Prédisposition génétique à une maladie , Étude d'association pangénomique , Glucokinase , Analyse de randomisation mendélienne , Humains , Protéines adaptatrices de la transduction du signal/génétique , Facteurs de risque , Appréciation des risques , Glycémie/métabolisme , Glucokinase/génétique , Glucokinase/métabolisme , Marqueurs biologiques/sang , Lipides/sang , Phénotype , Protéines de transport/génétique , Protéines de transport/métabolisme , Polymorphisme de nucléotide simple , Facteurs temps , Dyslipidémies/génétique , Dyslipidémies/sang , Dyslipidémies/diagnostic , Dyslipidémies/épidémiologie , Dyslipidémies/enzymologie , Stéatose hépatique/génétique , Stéatose hépatique/enzymologie , Stéatose hépatique/sang
10.
J Transl Med ; 22(1): 701, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39075482

RÉSUMÉ

BACKGROUND: The relationship between Helicobacter pylori (H. pylori) infection and metabolic dysfunction-associated steatotic liver disease (MASLD) has attracted increased clinical attention. However, most of those current studies involve cross-sectional studies and meta-analyses, and experimental mechanistic exploration still needs to be improved. This study aimed to investigate the mechanisms by which H. pylori impacts MASLD. METHODS: We established two H. pylori-infected (Cag A positive and Cag A negative) mouse models with 16 weeks of chow diet (CD) or high-fat diet (HFD) feeding. Body weight, liver triglyceride, blood glucose, serum biochemical parameters, inflammatory factors, and insulin resistance were measured, and histological analysis of liver tissues was performed. Mouse livers were subjected to transcriptome RNA sequencing analysis. RESULTS: Although H. pylori infection could not significantly affect serum inflammatory factor levels and serum biochemical parameters in mice, serum insulin and homeostatic model assessment for insulin resistance levels increased in CD mode. In contrast, H. pylori Cag A + infection significantly aggravated hepatic pathological steatosis induced by HFD and elevated serum inflammatory factors and lipid metabolism parameters. Hepatic transcriptomic analysis in the CD groups revealed 767 differentially expressed genes (DEGs) in the H. pylori Cag A + infected group and 1473 DEGs in the H. pylori Cag A- infected group, and the "nonalcoholic fatty liver disease" pathway was significantly enriched in KEGG analysis. There were 578 DEGs in H. pylori Cag A + infection combined with the HFD feeding group and 820 DEGs in the H. pylori Cag A- infected group. DEGs in the HFD groups were significantly enriched in "fatty acid degradation" and "PPAR pathway." Exploring the effect of different Cag A statuses on mouse liver revealed that fatty acid binding protein 5 was differentially expressed in Cag A- H. pylori. DEG enrichment pathways were concentrated in the "PPAR pathway" and "fatty acid degradation." CONCLUSIONS: Clinicians are expected to comprehend the impact of H. pylori on MASLD and better understand and manage MASLD. H. pylori infection may exacerbate the development of MASLD by regulating hepatic lipid metabolism, and the H. pylori virulence factor Cag A plays a vital role in this regulation.


Sujet(s)
Stéatose hépatique , Infections à Helicobacter , Helicobacter pylori , Métabolisme lipidique , Souris de lignée C57BL , Transcriptome , Animaux , Infections à Helicobacter/complications , Infections à Helicobacter/métabolisme , Métabolisme lipidique/génétique , Transcriptome/génétique , Stéatose hépatique/complications , Stéatose hépatique/microbiologie , Stéatose hépatique/métabolisme , Stéatose hépatique/génétique , Stéatose hépatique/anatomopathologie , Mâle , Alimentation riche en graisse , Foie/métabolisme , Foie/anatomopathologie , Insulinorésistance , Analyse de profil d'expression de gènes , Souris , Maladies métaboliques/microbiologie , Maladies métaboliques/complications , Maladies métaboliques/métabolisme , Maladies métaboliques/anatomopathologie , Maladies métaboliques/génétique , Voies et réseaux métaboliques/génétique
11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1869(7): 159532, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38981571

RÉSUMÉ

Hepatic adipogenesis has common mechanisms with adipocyte differentiation such as PPARγ involvement and the induction of adipose tissue-specific molecules. A previous report demonstrated that integrator complex subunit 6 (INTS6) is required for adipocyte differentiation. This study aimed to investigate INTS6 expression and its role in hepatic steatosis progression. The expression of INTS6 and PPARγ was examined in the liver of a mouse model of steatohepatitis and in paired liver biopsy samples from 11 patients with severe obesity and histologically proven metabolic dysfunction associated steatohepatitis (MASH) before and one year after bariatric surgery. To induce hepatocellular steatosis in vitro, an immortalized human hepatocyte cell line Hc3716 was treated with free fatty acids. In the steatohepatitis mouse model, we observed hepatic induction of INTS6, PPARγ, and adipocyte-specific genes. In contrast, ß-catenin which negatively regulates PPARγ was reduced. Biopsied human livers demonstrated a strong positive correlation (r2 = 0.8755) between INTS6 and PPARγ mRNA levels. After bariatric surgery, gene expressions of PPARγ, FABP4, and CD36 were mostly downregulated. In our in vitro experiments, we observed a concentration-dependent increase in Oil Red O staining in Hc3716 cells after treatment with the free fatty acids. Alongside this change, the expression of INTS6, PPARγ, and adipocyte-specific genes was induced. INTS6 knockdown using siRNA significantly suppressed cellular lipid accumulation together with induction of ß-catenin and PPARγ downregulation. Collectively, INTS6 expression closely correlates with PPARγ. INTS6 suppression significantly reduced hepatocyte steatosis via ß-catenin-PPARγ axis, indicating that INTS6 could be a novel therapeutic target for treating MASH.


Sujet(s)
Récepteur PPAR gamma , bêta-Caténine , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Humains , Animaux , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Souris , Mâle , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Stéatose hépatique/génétique , Femelle , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Lignée cellulaire , Souris de lignée C57BL , Modèles animaux de maladie humaine , Foie/métabolisme , Foie/anatomopathologie , Adulte d'âge moyen , Adulte , Protéines de liaison aux acides gras/métabolisme , Protéines de liaison aux acides gras/génétique , Antigènes CD36/métabolisme , Antigènes CD36/génétique
12.
Commun Biol ; 7(1): 897, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39048678

RÉSUMÉ

This study investigates sex-specific effects in a gain-of-function model to evaluate Nfil3 function in relation to high-fat diet (HFD)-induced metabolic dysfunction-associated steatotic liver disease (MASLD) and gut microbiota (GM)-induced alterations in the bile acid (BA) profile. MASLD is induced in both wild type and Nfil3-deficient (NKO) C57BL/6 J mice through an HFD. The hepatic immune response is evaluated using flow cytometry, revealing that NKO mice exhibit lower body weight, serum triglyceride (TG) levels, tissue injury, inflammation, and fat accumulation. The Nfil3 deletion reduces macrophage counts in fibrotic liver tissues, decreases proinflammatory gene and protein expression, and diminishes gut barrier function. Alpha and beta diversity analysis reveal increased GM alpha diversity across different sexes. The Nfil3 gene deletion modifies the BA profile, suggesting that negative feedback through the Nfil3-FXR-FGF15 axis facilitates BA recycling from the liver via enterohepatic circulation. Therefore, inhibiting Nfil3 in the liver offers a viable treatment approach for MASLD.


Sujet(s)
Alimentation riche en graisse , Souris de lignée C57BL , Souris knockout , Animaux , Souris , Mâle , Femelle , Alimentation riche en graisse/effets indésirables , Microbiome gastro-intestinal , Acides et sels biliaires/métabolisme , Foie/métabolisme , Foie/anatomopathologie , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/génétique , Stéatose hépatique non alcoolique/anatomopathologie , Modèles animaux de maladie humaine , Stéatose hépatique/métabolisme , Stéatose hépatique/génétique , Stéatose hépatique/anatomopathologie , Stéatose hépatique/étiologie , Facteurs de transcription à motif basique et à glissière à leucines
13.
Sci Rep ; 14(1): 15949, 2024 07 10.
Article de Anglais | MEDLINE | ID: mdl-38987612

RÉSUMÉ

Metabolic-associated steatohepatitis (MASH) and ulcerative colitis (UC) exhibit a complex interconnection with immune dysfunction, dysbiosis of the gut microbiota, and activation of inflammatory pathways. This study aims to identify and validate critical butyrate metabolism-related shared genes between both UC and MASH. Clinical information and gene expression profiles were sourced from the Gene Expression Omnibus (GEO) database. Shared butyrate metabolism-related differentially expressed genes (sBM-DEGs) between UC and MASH were identified via various bioinformatics methods. Functional enrichment analysis was performed, and UC patients were categorized into subtypes using the consensus clustering algorithm based on sBM-DEGs. Key genes within sBM-DEGs were screened through Random Forest, Support Vector Machines-Recursive Feature Elimination, and Light Gradient Boosting. The diagnostic efficacy of these genes was evaluated using receiver operating characteristic (ROC) analysis on independent datasets. Additionally, the expression levels of characteristic genes were validated across multiple independent datasets and human specimens. Forty-nine shared DEGs between UC and MASH were identified, with enrichment analysis highlighting significant involvement in immune, inflammatory, and metabolic pathways. The intersection of butyrate metabolism-related genes with these DEGs produced 10 sBM-DEGs. These genes facilitated the identification of molecular subtypes of UC patients using an unsupervised clustering approach. ANXA5, CD44, and SLC16A1 were pinpointed as hub genes through machine learning algorithms and feature importance rankings. ROC analysis confirmed their diagnostic efficacy in UC and MASH across various datasets. Additionally, the expression levels of these three hub genes showed significant correlations with immune cells. These findings were validated across independent datasets and human specimens, corroborating the bioinformatics analysis results. Integrated bioinformatics identified three significant biomarkers, ANXA5, CD44, and SLC16A1, as DEGs linked to butyrate metabolism. These findings offer new insights into the role of butyrate metabolism in the pathogenesis of UC and MASH, suggesting its potential as a valuable diagnostic biomarker.


Sujet(s)
Butyrates , Rectocolite hémorragique , Biologie informatique , Humains , Rectocolite hémorragique/génétique , Rectocolite hémorragique/métabolisme , Butyrates/métabolisme , Biologie informatique/méthodes , Analyse de profil d'expression de gènes , Courbe ROC , Stéatose hépatique/génétique , Stéatose hépatique/métabolisme , Bases de données génétiques , Transcriptome , Microbiome gastro-intestinal/génétique
14.
FASEB J ; 38(11): e23717, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38837270

RÉSUMÉ

Selenoprotein I (Selenoi) is highly expressed in liver and plays a key role in lipid metabolism as a phosphatidylethanolamine (PE) synthase. However, the precise function of Selenoi in the liver remains elusive. In the study, we generated hepatocyte-specific Selenoi conditional knockout (cKO) mice on a high-fat diet to identify the physiological function of Selenoi. The cKO group exhibited a significant increase in body weight, with a 15.6% and 13.7% increase in fat accumulation in white adipose tissue (WAT) and the liver, respectively. Downregulation of the lipolysis-related protein (p-Hsl) and upregulation of the adipogenesis-related protein (Fasn) were observed in the liver of cKO mice. The cKO group also showed decreased oxygen consumption (VO2), carbon dioxide production (VCO2), and energy expenditure (p < .05). Moreover, various metabolites of the steroid hormone synthesis pathway were affected in the liver of cKO mice. A potential cascade of Selenoi-phosphatidylethanolamine-steroid hormone synthesis might serve as a core mechanism that links hepatocyte-specific Selenoi cKO to biochemical and molecular reactions. In conclusion, we revealed that Selenoi inhibits body fat accumulation and hepatic steatosis and elevates energy consumption; this protein could also be considered a therapeutic target for such related diseases.


Sujet(s)
Stéatose hépatique , Hépatocytes , Souris knockout , Obésité , Animaux , Souris , Obésité/métabolisme , Obésité/génétique , Obésité/étiologie , Hépatocytes/métabolisme , Stéatose hépatique/métabolisme , Stéatose hépatique/étiologie , Stéatose hépatique/génétique , Stéatose hépatique/anatomopathologie , Sélénoprotéines/métabolisme , Sélénoprotéines/génétique , Alimentation riche en graisse/effets indésirables , Mâle , Foie/métabolisme , Métabolisme énergétique , Métabolisme lipidique , Souris de lignée C57BL , Tissu adipeux blanc/métabolisme
15.
Nutrients ; 16(12)2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38931155

RÉSUMÉ

Gut microbiota might affect the severity and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). We aimed to characterize gut dysbiosis and clinical parameters regarding fibrosis stages assessed by magnetic resonance elastography. This study included 156 patients with MASLD, stratified into no/mild fibrosis (F0-F1) and moderate/severe fibrosis (F2-F4). Fecal specimens were sequenced targeting the V4 region of the 16S rRNA gene and analyzed using bioinformatics. The genotyping of PNPLA3, TM6SF2, and HSD17B13 was assessed by allelic discrimination assays. Our data showed that gut microbial profiles between groups significantly differed in beta-diversity but not in alpha-diversity indices. Enriched Fusobacterium and Escherichia_Shigella, and depleted Lachnospira were found in the F2-F4 group versus the F0-F1 group. Compared to F0-F1, the F2-F4 group had elevated plasma surrogate markers of gut epithelial permeability and bacterial translocation. The bacterial genera, PNPLA3 polymorphisms, old age, and diabetes were independently associated with advanced fibrosis in multivariable analyses. Using the Random Forest classifier, the gut microbial signature of three genera could differentiate the groups with high diagnostic accuracy (AUC of 0.93). These results indicated that the imbalance of enriched pathogenic genera and decreased beneficial bacteria, in association with several clinical and genetic factors, were potential contributors to the pathogenesis and progression of MASLD.


Sujet(s)
Microbiome gastro-intestinal , Cirrhose du foie , Protéines membranaires , Indice de gravité de la maladie , Humains , Microbiome gastro-intestinal/génétique , Cirrhose du foie/microbiologie , Cirrhose du foie/génétique , Femelle , Mâle , Adulte d'âge moyen , Protéines membranaires/génétique , Triacylglycerol lipase/génétique , Sujet âgé , ARN ribosomique 16S/génétique , Dysbiose , Stéatose hépatique/microbiologie , Stéatose hépatique/génétique , Fèces/microbiologie , Adulte , Variation génétique , Imagerie d'élasticité tissulaire , Bactéries/génétique , Bactéries/isolement et purification , Bactéries/classification , Acyltransferases , 17-Hydroxysteroid dehydrogenases , Calcium-independent phospholipase A2
16.
Viruses ; 16(6)2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38932276

RÉSUMÉ

Metabolic-associated fatty liver disease (MAFLD) is a risk factor for severe COVID-19. This study explores the potential influence of gut hormone receptor and immune response gene expression on COVID-19 outcomes in MAFLD patients. METHODS: We investigated gene expression levels of AHR, FFAR2, FXR, and TGR5 in patients with MAFLD and COVID-19 compared to controls. We examined associations between gene expression and clinical outcomes. RESULTS: COVID-19 patients displayed altered AHR expression, potentially impacting immune response and recovery. Downregulated AHR in patients with MAFLD correlated with increased coagulation parameters. Elevated FFAR2 expression in patients with MAFLD was linked to specific immune cell populations and hospital stay duration. A significantly lower FXR expression was observed in both MAFLD and severe COVID-19. CONCLUSION: Our findings suggest potential modulatory roles for AHR, FFAR2, and FXR in COVID-19 and MAFLD.


Sujet(s)
COVID-19 , Récepteurs à hydrocarbure aromatique , Récepteurs couplés aux protéines G , SARS-CoV-2 , Humains , COVID-19/génétique , COVID-19/virologie , COVID-19/immunologie , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme , Mâle , Femelle , Récepteurs à hydrocarbure aromatique/génétique , Récepteurs à hydrocarbure aromatique/métabolisme , Adulte d'âge moyen , Sujet âgé , Récepteurs cytoplasmiques et nucléaires/métabolisme , Récepteurs cytoplasmiques et nucléaires/génétique , Expression des gènes , Stéatose hépatique/génétique , Stéatose hépatique/virologie , Adulte , Protéines de liaison à l'ARN , Facteurs de transcription à motif basique hélice-boucle-hélice
17.
BMC Med ; 22(1): 270, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38926684

RÉSUMÉ

BACKGROUND: Previous studies, including Mendelian randomization (MR), have demonstrated type 2 diabetes (T2D) and glycemic traits are associated with increased risk of metabolic dysfunction-associated steatotic liver disease (MASLD). However, few studies have explored the underlying pathway, such as the role of iron homeostasis. METHODS: We used a two-step MR approach to investigate the associations of genetic liability to T2D, glycemic traits, iron biomarkers, and liver diseases. We analyzed summary statistics from various genome-wide association studies of T2D (n = 933,970), glycemic traits (n ≤ 209,605), iron biomarkers (n ≤ 246,139), MASLD (n ≤ 972,707), and related biomarkers (alanine aminotransferase (ALT) and proton density fat fraction (PDFF)). Our primary analysis was based on inverse-variance weighting, followed by several sensitivity analyses. We also conducted mediation analyses and explored the role of liver iron in post hoc analysis. RESULTS: Genetic liability to T2D and elevated fasting insulin (FI) likely increased risk of liver steatosis (ORliability to T2D: 1.14 per doubling in the prevalence, 95% CI: 1.10, 1.19; ORFI: 3.31 per log pmol/l, 95% CI: 1.92, 5.72) and related biomarkers. Liability to T2D also likely increased the risk of developing liver cirrhosis. Genetically elevated ferritin, serum iron, and liver iron were associated with higher risk of liver steatosis (ORferritin: 1.25 per SD, 95% CI 1.07, 1.46; ORliver iron: 1.15 per SD, 95% CI: 1.05, 1.26) and liver cirrhosis (ORserum iron: 1.31, 95% CI: 1.06, 1.63; ORliver iron: 1.34, 95% CI: 1.07, 1.68). Ferritin partially mediated the association between FI and liver steatosis (proportion mediated: 7%, 95% CI: 2-12%). CONCLUSIONS: Our study provides credible evidence on the causal role of T2D and elevated insulin in liver steatosis and cirrhosis risk and indicates ferritin may play a mediating role in this association.


Sujet(s)
Marqueurs biologiques , Diabète de type 2 , Homéostasie , Fer , Cirrhose du foie , Analyse de randomisation mendélienne , Humains , Diabète de type 2/génétique , Fer/sang , Fer/métabolisme , Marqueurs biologiques/sang , Cirrhose du foie/génétique , Stéatose hépatique/génétique , Étude d'association pangénomique , Glycémie/métabolisme
18.
Nat Commun ; 15(1): 4847, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38844467

RÉSUMÉ

The I148M variant of PNPLA3 is closely associated with hepatic steatosis. Recent evidence indicates that the I148M mutant functions as an inhibitor of PNPLA2/ATGL-mediated lipolysis, leaving the role of wild-type PNPLA3 undefined. Despite showing a triglyceride hydrolase activity in vitro, PNPLA3 has yet to be established as a lipase in vivo. Here, we show that PNPLA3 preferentially hydrolyzes polyunsaturated triglycerides, mobilizing polyunsaturated fatty acids for phospholipid desaturation and enhancing hepatic secretion of triglyceride-rich lipoproteins. Under lipogenic conditions, mice with liver-specific knockout or acute knockdown of PNPLA3 exhibit aggravated liver steatosis and reduced plasma VLDL-triglyceride levels. Similarly, I148M-knockin mice show decreased hepatic triglyceride secretion during lipogenic stimulation. Our results highlight a specific context whereby the wild-type PNPLA3 facilitates the balance between hepatic triglyceride storage and secretion, and suggest the potential contribution of a loss-of-function by the I148M variant to the development of fatty liver disease in humans.


Sujet(s)
Acides gras insaturés , Triacylglycerol lipase , Lipoprotéines VLDL , Foie , Souris knockout , Triglycéride , Animaux , Triacylglycerol lipase/métabolisme , Triacylglycerol lipase/génétique , Foie/métabolisme , Triglycéride/métabolisme , Souris , Lipoprotéines VLDL/métabolisme , Humains , Acides gras insaturés/métabolisme , Mâle , Stéatose hépatique/métabolisme , Stéatose hépatique/génétique , Souris de lignée C57BL , Lipolyse , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Acyltransferases , Calcium-independent phospholipase A2
19.
Hepatol Commun ; 8(6)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38836837

RÉSUMÉ

BACKGROUND: Abnormal phospholipid metabolism is linked to metabolic dysfunction-associated steatotic liver disease (MASLD) development and progression. We aimed to clarify whether genetic variants of phospholipid metabolism modify these relationships. METHODS: This case-control study consecutively recruited 600 patients who underwent MRI-based proton density fat fraction examination (240 participants with serum metabonomics analysis, 128 biopsy-proven cases) as 3 groups: healthy control, nonobese MASLD, and obese MASLD, (n = 200 cases each). Ten variants of phospholipid metabolism-related genes [phospholipase A2 Group VII rs1805018, rs76863441, rs1421378, and rs1051931; phospholipase A2 receptor 1 (PLA2R1) rs35771982, rs3828323, and rs3749117; paraoxonase-1 rs662 and rs854560; and ceramide synthase 4 (CERS4) rs17160348)] were genotyped using SNaPshot. RESULTS: The T-allele of CERS4 rs17160348 was associated with a higher risk of both obese and nonobese MASLD (OR: 1.95, 95% CI: 1.20-3.15; OR: 1.76, 95% CI: 1.08-2.86, respectively). PLA2R1 rs35771982-allele is a risk factor for nonobese MASLD (OR: 1.66, 95% CI: 1.11-1.24), moderate-to-severe steatosis (OR: 3.24, 95% CI: 1.96-6.22), and steatohepatitis (OR: 2.61, 95% CI: 1.15-3.87), while the paraoxonase-1 rs854560 T-allele (OR: 0.50, 95% CI: 0.26-0.97) and PLA2R1 rs3749117 C-allele (OR: 1.70, 95% CI: 1.14-2.52) are closely related to obese MASLD. After adjusting for sphingomyelin level, the effect of the PLA2R1 rs35771982CC allele on MASLD was attenuated. Furthermore, similar effects on the association between the CERS4 rs17160348 C allele and MASLD were observed for phosphatidylcholine, phosphatidic acid, sphingomyelin, and phosphatidylinositol. CONCLUSIONS: The mutations in PLA2R1 rs35771982 and CERS4 rs17160348 presented detrimental impact on the risk of occurrence and disease severity in nonobese MASLD through altered phospholipid metabolism.


Sujet(s)
Génotype , Récepteurs à la phospholipase A2 , Humains , Mâle , Femelle , Adulte d'âge moyen , Études cas-témoins , Récepteurs à la phospholipase A2/génétique , Phospholipides/sang , Adulte , Obésité/génétique , Polymorphisme de nucléotide simple , Stéatose hépatique/génétique , Prédisposition génétique à une maladie/génétique
20.
Mol Metab ; 86: 101977, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38936659

RÉSUMÉ

OBJECTIVE: The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) continues to rise with the increasing obesity epidemic. Rezdiffra as an activator of a thyroid hormone receptor-beta is the only Food and Drug Administration approved therapy. As such, there is a critical need to improve our understanding of gene expression regulation and signaling transduction in MASLD to develop new therapies. Matrin-3 is a DNA- and RNA-binding protein involved in the pathogenesis of human diseases. Here we examined its previously uncharacterized role in limiting hepatic steatosis and stress response via the constitutive androstane receptor (CAR). METHODS: Matrin-3 floxed and liver-specific knockout mice were fed either a chow diet or 60 kcal% high-fat diet (HFD) for up to 16 weeks. The mice were euthanized for different analysis including liver histology, lipid levels, and gene expression. Bulk RNA-seq, bulk ATAC-seq, and single-nucleus Multiome were used to examine changes of transcriptome and chromatin accessibility in the liver. Integrative bioinformatics analysis of our data and publicly available datasets and different biochemical assays were performed to identify underlying the molecular mechanisms mediating matrin-3's effects. Liver-tropic adeno-associated virus was used to restore the expression of CAR for lipid, acute phase genes, and histological analysis. RESULTS: Matrin-3 expression is induced in the steatotic livers of mice. Liver-specific matrin-3 deletion exacerbated HFD-induced steatosis, acute phase response, and inflammation in the liver of female mice. The transcriptome and chromatin accessibility were re-programmed in the liver of these mice with signatures indicating that CAR signaling is dysregulated. Mechanistically, matrin-3 interacts with CAR mRNA, and matrin-3 deficiency promotes CAR mRNA degradation. Consequently, matrin-3 deletion impaired CAR signaling by reducing CAR expression. Matrin-3 levels positively correlate with CAR expression in human livers. Ces2a and Il1r1 were identified as new target genes of CAR. Interestingly, we found that CAR discords with the expression of its target genes including Cyp2b10 and Ces2a in response to HFD, indicating CAR signaling is dysregulated by HFD despite increased CAR expression. Dysregulated CAR signaling upon matrin-3 deficiency reduced Ces2a and de-repressed Il1r1 expression. CAR restoration partially abrogated the dysregulated gene expression, exacerbated hepatic steatosis, acute phase response, and inflammation in liver-specific matrin-3 knockout mice fed a HFD. CONCLUSIONS: Our findings demonstrate that matrin-3 is a key upstream regulator maintaining CAR signaling upon metabolic stress, and the matrin-3-CAR axis limits hepatic steatosis and stress response signaling that may give insights for therapeutic intervention.


Sujet(s)
Récepteur constitutif des androstanes , Stéatose hépatique , Foie , Souris knockout , Animaux , Souris , Foie/métabolisme , Stéatose hépatique/métabolisme , Stéatose hépatique/génétique , Alimentation riche en graisse/effets indésirables , Mâle , Souris de lignée C57BL , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/génétique , Transduction du signal , Stress physiologique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE