Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.655
Filtrer
1.
J Nutr Sci Vitaminol (Tokyo) ; 70(4): 318-327, 2024.
Article de Anglais | MEDLINE | ID: mdl-39218693

RÉSUMÉ

Recently, we have demonstrated that mice, cultured embryos in α-minimum essential medium (αMEM) and subsequent fed a high-fat, high-sugar diet, developed steatohepatitis. In this study, we investigated using these samples whether the expression of lipid droplet formation genes in the liver is higher in MEM mice, whether these expressions are regulated by histone acetylation, writers/readers of histone acetylation, and the transcriptional factors of endoplasmic reticulum stress. Mice were produced by two-cell embryos in αMEM or standard potassium simplex-optimized medium (control) in vitro for 48 h, and implanted into an oviduct for spontaneous delivery. MEM and control-mice were fed a high-fat, high-sugar diet for 18 wk, and then liver samples were collected and analyzed by histology, qRT-PCR, and chromatin immunoprecipitation assay. Gene expression of Cidea, Cidec, and Plin4 were higher in MEM mice and histone H3K9 acetylation, BRD4, and CBP were higher in MEM mice than in control mice around those genes. However, the binding of endoplasmic reticulum stress-related transcription factors (ATF4, CHOP and C/EBPα) around those genes in the liver, was not clearly differed between MEM mice and control mice. The increased expression of Cidea, Cidec and Plin4 in the liver, accompanied by the development of steatohepatitis in mice induced is positively associated with increased histone H3K9 acetylation and CBP and BRD4 binding around these genes.


Sujet(s)
Stress du réticulum endoplasmique , Stéatose hépatique , Histone , Gouttelettes lipidiques , Foie , Animaux , Histone/métabolisme , Acétylation , Gouttelettes lipidiques/métabolisme , Souris , Femelle , Foie/métabolisme , Stéatose hépatique/métabolisme , Stéatose hépatique/génétique , Stéatose hépatique/étiologie , Facteur de transcription ATF-4/métabolisme , Facteur de transcription ATF-4/génétique , Alimentation riche en graisse/effets indésirables , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Facteur de transcription CHOP/métabolisme , Facteur de transcription CHOP/génétique , Protéines liant les séquences stimulatrices de type CCAAT/métabolisme , Protéines liant les séquences stimulatrices de type CCAAT/génétique , Protéines régulatrices de l'apoptose/métabolisme , Protéines régulatrices de l'apoptose/génétique
2.
Int J Mol Sci ; 25(16)2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-39201418

RÉSUMÉ

Type 2 diabetes mellitus (T2DM), often featuring hyperglycemia or insulin resistance, is a global health concern that is increasing in prevalence in the United States and worldwide. A common complication is metabolic dysfunction-associated steatotic liver disease (MASLD), the hepatic manifestation of metabolic syndrome that is also rapidly increasing in prevalence. The majority of patients with T2DM will experience MASLD, and likewise, individuals with MASLD are at an increased risk for developing T2DM. These two disorders may act synergistically, in part due to increased lipotoxicity and inflammation within the liver, among other causes. However, the pathophysiological mechanisms by which this occurs are unclear, as is how the improvement of one disorder can ameliorate the other. This review aims to discuss the pathogenic interactions between T2D and MASLD, and will highlight novel therapeutic targets and ongoing clinical trials for the treatment of these diseases.


Sujet(s)
Diabète de type 2 , Humains , Diabète de type 2/métabolisme , Diabète de type 2/complications , Animaux , Syndrome métabolique X/métabolisme , Syndrome métabolique X/complications , Insulinorésistance , Stéatose hépatique/métabolisme , Stéatose hépatique/étiologie , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/complications
3.
Int J Mol Sci ; 25(16)2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39201759

RÉSUMÉ

Inhibiting diacylglycerol acetyltransferase (DGAT1, DGAT2) enzymes (iDGAT1, iDGAT2), involved in triglyceride (TG) synthesis, improves hepatic steatosis in metabolic dysfunction-associated steatotic liver disease (MASLD) patients. However, their potential synergism in disease onset (SLD) and progression (metabolic dysfunction-associated steatohepatitis, fibrosis) has been poorly explored. We investigated iDGAT1 and iDGAT2 efficacy, alone or combined (iDGAT1/2) on fat accumulation and hepatocellular injury in hepatocytes (HepG2) and on fibrogenic processes in hepatic stellate cells (LX2). We further tested whether the addition of MitoQ antioxidant to iDGAT1/2 would enhance their effects. SLD and MASH conditions were reproduced in vitro by supplementing Dulbecco's Modified Eagle's Medium (DMEM) with palmitic/oleic acids (PAOA) alone (SLD-medium), or plus Lipopolisaccaride (LPS), fructose, and glucose (MASH-medium). In SLD-medium, iDGAT1 and iDGAT2 individually, and even more in combination, reduced TG synthesis in HepG2 cells. Markers of hepatocellular damage were slightly decreased after single iDGAT exposure. Conversely, iDGAT1/2 counteracted ER/oxidative stress and inflammation and enhanced mitochondrial Tricarboxylic acid cycle (TCA) and respiration. In HepG2 cells under a MASH-like condition, only iDGAT1/2 effectively ameliorated TG content and oxidative and inflammatory mediators, further improving bioenergetic balance. LX2 cells, challenged with SLD/MASH media, showed less proliferation and slower migration rates in response to iDGAT1/2 drugs. MitoQ combined with iDGAT1/2 improved cell viability and dampened free fatty acid release by stimulating ß-oxidation. Dual DGAT inhibition combined with antioxidants open new perspectives for MASLD management.


Sujet(s)
Diacylglycerol O-acyltransferase , Triglycéride , Humains , Diacylglycerol O-acyltransferase/métabolisme , Diacylglycerol O-acyltransferase/antagonistes et inhibiteurs , Cellules HepG2 , Triglycéride/métabolisme , Antienzymes/pharmacologie , Antienzymes/usage thérapeutique , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Stéatose hépatique/traitement médicamenteux , Stéatose hépatique/métabolisme , Stéatose hépatique/étiologie , Stress oxydatif/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques
4.
Nutrients ; 16(16)2024 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-39203941

RÉSUMÉ

Both maternal obesity and postnatal consumption of obesogenic diets contribute to the development of metabolic dysfunction-associated steatotic liver disease (MASLD) and hepatocellular carcinoma (HCC). However, there is no consensus as to whether diets that are high in fat or carbohydrates/sugars differentially influence the development of HCC. Moreover, the long-term effects of prenatal HF exposure on HCC and whether this is influenced by postnatal diet has not yet been evaluated. C57BL/6 dams were fed either a low-fat, high-carbohydrate control (C) or low-carbohydrate, high-fat (HF) diet. At weaning, male and female offspring were fed the C or HF diet, generating four diet groups: C/C, C/HF, HF/C and HF/HF. Tissues were collected at 16 months of age and livers were assessed for MASLD and HCC. Glucose regulation and pancreatic morphology were also evaluated. Liver tissues were assessed for markers of glycolysis and fatty acid metabolism and validated using a human HCC bioinformatic database. Both C/HF and HF/HF mice developed obesity, hyperinsulinemia and a greater degree of MASLD than C/C and HF/C offspring. However, despite significant liver and pancreas pathology, C/HF mice had the lowest incidence of HCC while tumour burden was highest in HF/C male offspring. The molecular profile of HCC mouse samples suggested an upregulation of the pentose phosphate pathway and a downregulation of fatty acid synthesis and oxidation, which was largely validated in the human dataset. Both pre-weaning HF diet exposure and post-weaning consumption of a high-carbohydrate diet increased the risk of developing spontaneous HCC in aged mice. However, the influence of pre-weaning HF feeding on HCC development appeared to be stronger in the context of post-weaning obesity. As rates of maternal obesity continue to rise, this has implications for the future incidence of HCC and possible dietary manipulation of offspring carbohydrate intake to counteract this risk.


Sujet(s)
Carcinome hépatocellulaire , Alimentation riche en graisse , Tumeurs du foie , Souris de lignée C57BL , Sevrage , Animaux , Femelle , Alimentation riche en graisse/effets indésirables , Carcinome hépatocellulaire/étiologie , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/étiologie , Tumeurs du foie/métabolisme , Grossesse , Mâle , Souris , Effets différés de l'exposition prénatale à des facteurs de risque , Phénomènes physiologiques nutritionnels maternels , Foie/métabolisme , Foie/anatomopathologie , Obésité , Stéatose hépatique/étiologie , Hydrates de carbone alimentaires/effets indésirables , Hydrates de carbone alimentaires/administration et posologie
5.
Sci Rep ; 14(1): 19292, 2024 08 20.
Article de Anglais | MEDLINE | ID: mdl-39164313

RÉSUMÉ

Metabolic dysfunction-associated fatty liver disease (MAFLD) is a disease that causes an abnormal accumulation of fat in the liver, triggering inflammation and fibrosis, the mechanism of which is not fully understood and for which there is a lack of specific drug therapy. Far-infrared radiation (FIR) has demonstrated evident therapeutic efficacy across various diseases, and novel nanomaterial graphene patches can emit it through electric heating. This study aimed to investigate the potential protective effects of FIR against MAFLD. Mice were fed with a MCD diet to mimic MAFLD progression, and histopathology analysis, biochemical analysis, RT-qPCR, and Western blotting analysis were performed to assess the effect of FIR on MAFLD in vivo. The effect of FIR treatment on MAFLD in vitro was investigated by biochemical analysis and gene expression profiling of hepatocytes. Mice subjected to the MCD diet and treated with FIR exhibited reduced hepatic lipid deposition, inflammation, fibrosis and liver damage. The therapeutic effect exerted by FIR in mice may be caused by the enhancement of AMPK phosphorylation and inhibition of the TGFß1-SMAD2/3 pathway. Besides, FIR intervention alleviated MAFLD in hepatocytes in vitro and the results were verified by gene expression profiling. Our results revealed a promising potential of FIR as a novel therapeutic approach for MAFLD.


Sujet(s)
Hépatocytes , Rayons infrarouges , Cirrhose du foie , Animaux , Souris , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Cirrhose du foie/étiologie , Hépatocytes/métabolisme , Mâle , Facteur de croissance transformant bêta-1/métabolisme , Souris de lignée C57BL , Modèles animaux de maladie humaine , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Stéatose hépatique/étiologie , Foie/métabolisme , Foie/anatomopathologie , Foie/effets des radiations , Transduction du signal , Protéine Smad-3/métabolisme , Protéine Smad2/métabolisme , Phosphorylation
6.
Sci Rep ; 14(1): 18006, 2024 08 03.
Article de Anglais | MEDLINE | ID: mdl-39097615

RÉSUMÉ

Choline is recognized as an essential nutrient for Atlantic salmon at all developmental stages. However, its dietary requirement is not well defined. Choline plays a critical role in lipid transport, and the clearest deficiency sign is intestinal steatosis. The present work, aiming to find whether lipid source and fish size may affect steatosis symptoms, was one of a series of studies conducted to identify which production-related conditions may influence choline requirement. Six choline-deficient diets were formulated varying in ratios of rapeseed oil to fish oil and fed to Atlantic salmon of 1.5 and 4.5 kg. After eight weeks, somatic characteristics were observed, and the severity of intestinal steatosis was assessed by histological, biochemical, and molecular analyses. Fatty acid composition in pyloric intestine, mesenteric tissue, and liver samples was also quantified. The increasing rapeseed oil level increased lipid digestibility markedly, enhancing lipid supply to the fish. Moreover, small fish consumed more feed, and consequently had a higher lipid intake. In conclusion, the results showed that choline requirement depends on dietary lipid load, which depends on the fatty acid profile as well as the fish size.


Sujet(s)
Aliment pour animaux , Huiles de poisson , Huile de colza , Salmo salar , Animaux , Huile de colza/administration et posologie , Salmo salar/métabolisme , Salmo salar/croissance et développement , Huiles de poisson/administration et posologie , Aliment pour animaux/analyse , Acides gras/métabolisme , Acides gras/analyse , Maladies des poissons/anatomopathologie , Maladies des poissons/métabolisme , Stéatose hépatique/médecine vétérinaire , Stéatose hépatique/métabolisme , Stéatose hépatique/étiologie , Stéatose hépatique/anatomopathologie , Choline/métabolisme , Choline/administration et posologie , Régime alimentaire/médecine vétérinaire , Foie/métabolisme , Foie/anatomopathologie
7.
Int J Mol Sci ; 25(15)2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-39125804

RÉSUMÉ

Obesity is an emerging public health problem. Chronic low-grade inflammation is considered a major promotor of obesity-induced secondary diseases such as cardiovascular and fatty liver disease, type 2 diabetes mellitus, and several cancer entities. Most preliminary studies on obesity-induced immune responses have been conducted in male rodents. Sex-specific differences between men and women in obesity-induced immune dysregulation have not yet been fully outlined but are highly relevant to optimizing prevention strategies for overweight-associated complications. In this study, we fed C57BL/6 female vs. male mice with either standard chow or an obesity-inducing diet (OD). Blood and spleen immune cells were isolated and analyzed by flow cytometry. Lean control mice showed no sex bias in systemic and splenic immune cell composition, whereas the immune responses to obesity were significantly distinct between female and male mice. While immune cell alterations in male OD mice were characterized by a significant reduction in T cells and an increase in myeloid-derived suppressor cells (MDSC), female OD mice displayed preserved T cell numbers. The sex-dependent differences in obesity-induced T cell dysregulation were associated with varying susceptibility to body weight gain and fatty liver disease: Male mice showed significantly more hepatic inflammation and histopathological stigmata of fatty liver in comparison to female OD mice. Our findings indicate that sex impacts susceptibility to obesity-induced T cell dysregulation, which might explain sex-dependent different incidences in the development of obesity-associated secondary diseases. These results provide novel insights into the understanding of obesity-induced chronic inflammation from a sex-specific perspective. Given that most nutrition, exercise, and therapeutic recommendations for the prevention of obesity-associated comorbidities do not differentiate between men and women, the data of this study are clinically relevant and should be taken into consideration in future trials and treatment strategies.


Sujet(s)
Souris de lignée C57BL , Obésité , Lymphocytes T , Animaux , Obésité/immunologie , Obésité/complications , Obésité/étiologie , Femelle , Mâle , Souris , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Alimentation riche en graisse/effets indésirables , Facteurs sexuels , Rate/immunologie , Rate/anatomopathologie , Caractères sexuels , Cellules myéloïdes suppressives/immunologie , Cellules myéloïdes suppressives/métabolisme , Stéatose hépatique/étiologie , Stéatose hépatique/immunologie , Stéatose hépatique/anatomopathologie , Inflammation/immunologie , Inflammation/anatomopathologie , Inflammation/étiologie
8.
Clin Transplant ; 38(8): e15437, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39171566

RÉSUMÉ

BACKGROUND AND AIMS: Biopsy-proven severe graft steatosis is associated with adverse outcomes after liver transplantation. The concomitant presence of metabolic risk factors might further increase this risk. We studied the association between graft steatosis and metabolic risk factors in the donor, with recipient outcomes after liver transplantation. METHODS: We analyzed data from all consecutive first adult full-graft donation after brain death (DBD) liver transplantations performed in the Eurotransplant region between 2010 and 2020. The presence of graft steatosis and metabolic risk factors was assessed through a review of donor (imaging) reports, and associations with recipient retransplantation-free survival were studied through survival analyses. RESULTS: Of 12 174 transplantations, graft steatosis was detected in 2689 (22.1%), and donor diabetes mellitus (DM), hypertension, and dyslipidemia were present in 1245 (10.2%), 5056 (41.5%), and 524 (4.3%). In multivariable Cox regression analysis, graft steatosis (adjusted HR [aHR] 1.197, p < 0.001) and donor DM (aHR 1.157, p = 0.004) were independently associated with impaired retransplantation-free survival. Graft steatosis and donor DM conferred an additive risk of retransplantation or death (DM alone, aHR: 1.156 [p = 0.0185]; steatosis alone, aHR: 1.200 [p < 0.001]; both steatosis and DM, aHR: 1.381 [p < 0.001]). Findings were consistent in sensitivity analyses focusing on retransplantation-free survival within 7 days. CONCLUSIONS: Graft steatosis and donor diabetes mellitus additively increase the risk of retransplantation or death in adult DBD liver transplantation. Future studies should focus on methods to assess and improve the quality of these high-risk grafts. Until such time, caution should be exercised when considering these grafts for transplantation.


Sujet(s)
Stéatose hépatique , Survie du greffon , Transplantation hépatique , Complications postopératoires , Enregistrements , Donneurs de tissus , Humains , Femelle , Mâle , Transplantation hépatique/effets indésirables , Transplantation hépatique/mortalité , Adulte d'âge moyen , Stéatose hépatique/anatomopathologie , Stéatose hépatique/étiologie , Stéatose hépatique/complications , Stéatose hépatique/chirurgie , Donneurs de tissus/ressources et distribution , Facteurs de risque , Études de suivi , Pronostic , Adulte , Europe/épidémiologie , Taux de survie , Diabète , Rejet du greffon/étiologie , Rejet du greffon/mortalité , Études rétrospectives , Receveurs de transplantation/statistiques et données numériques
9.
Ann N Y Acad Sci ; 1538(1): 21-33, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38996214

RÉSUMÉ

Metabolic dysfunction-associated liver disease (MASLD) and steatohepatitis (MASH) are becoming the most common causes of chronic liver disease in the United States and worldwide due to the obesity and diabetes epidemics. It is estimated that by 2030 close to 100 million people might be affected and patients with type 2 diabetes are especially at high risk. Twenty to 30% of patients with MASLD can progress to MASH, which is characterized by steatosis, necroinflammation, hepatocyte ballooning, and in advanced cases, fibrosis progressing to cirrhosis. Clinically, it is recognized that disease progression in diabetic patients is accelerated and the role of various genetic and epigenetic factors, as well as cell-matrix interactions in fibrosis and stromal remodeling, have recently been recognized. While there has been great progress in drug development and clinical trials for MASLD/MASH, the complexity of these pathways highlights the need to improve diagnosis/early detection and develop more successful antifibrotic therapies that not only prevent but reverse fibrosis.


Sujet(s)
Diabète de type 2 , Cirrhose du foie , Humains , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Diabète de type 2/métabolisme , Diabète de type 2/complications , Évolution de la maladie , Animaux , Matrice extracellulaire/métabolisme , Stéatose hépatique/métabolisme , Stéatose hépatique/étiologie
10.
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
11.
Sci Rep ; 14(1): 17558, 2024 07 30.
Article de Anglais | MEDLINE | ID: mdl-39080285

RÉSUMÉ

Metabolic dysfunction-associated steatotic liver disease (MASLD) and related steatohepatitis (MASH) are common among obese patients and may improve after metabolic/bariatric surgery (MBS). 93 Patients undergoing MBS in 2021-2022 were prospectively enrolled. Liver stiffness measurement (LSM; via vibration-controlled transient elastography [VCTE], point [pSWE] and 2D [2DSWE] shear wave elastography) and non-invasive steatosis assessment (via controlled attenuation parameter [CAP]) were performed before (baseline [BL]) and three months (M3) after surgery. 93 patients (median age 40.9 years, 68.8% female, median BL-BMI: 46.0 kg/m2) were included. BL-liver biopsy showed MASLD in 82.8% and MASH in 34.4% of patients. At M3 the median relative total weight loss (%TWL) was 20.1% and the median BMI was 36.1 kg/m2. LSM assessed by VCTE and 2DSWE, as well as median CAP all decreased significantly from BL to M3 both in the overall cohort and among patients with MASH. There was a decrease from BL to M3 in median levels of ALT (34.0 U/L to 31 U/L; p = 0.025), gamma glutamyl transferase (BL: 30.0 to 21.0 U/L; p < 0.001) and MASLD fibrosis score (BL: - 0.97 to - 1.74; p < 0.001). Decreasing LSM and CAP, as well as liver injury markers suggest an improvement of MASLD/MASH as early as 3 months after MBS.


Sujet(s)
Chirurgie bariatrique , Imagerie d'élasticité tissulaire , Stéatose hépatique , Foie , Humains , Femelle , Chirurgie bariatrique/méthodes , Mâle , Adulte , Études prospectives , Stéatose hépatique/chirurgie , Stéatose hépatique/métabolisme , Stéatose hépatique/étiologie , Foie/métabolisme , Foie/anatomopathologie , Foie/imagerie diagnostique , Foie/chirurgie , Adulte d'âge moyen , Obésité/chirurgie , Obésité/complications , Obésité/métabolisme , Résultat thérapeutique
12.
FASEB J ; 38(13): e23788, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38963329

RÉSUMÉ

Intermittent hypoxia (IH) is an independent risk factor for metabolic dysfunction-associated fatty liver disease (MAFLD). Copper deficiency can disrupt redox homeostasis, iron, and lipid metabolism. Here, we investigated whether hepatic copper deficiency plays a role in IH-associated MAFLD and explored the underlying mechanism(s). Male C57BL/6 mice were fed a western-type diet with adequate copper (CuA) or marginally deficient copper (CuD) and were exposed separately to room air (RA) or IH. Hepatic histology, plasma biomarkers, copper-iron status, and oxidative stress were assessed. An in vitro HepG2 cell lipotoxicity model and proteomic analysis were used to elucidate the specific targets involved. We observed that there were no differences in hepatic phenotypes between CuA-fed and CuD-fed mice under RA. However, in IH exposure, CuD-fed mice showed more pronounced hepatic steatosis, liver injury, and oxidative stress than CuA-fed mice. IH induced copper accumulation in the brain and heart and exacerbated hepatic copper deficiency and secondary iron deposition. In vitro, CuD-treated cells with IH exposure showed elevated levels of lipid accumulation, oxidative stress, and ferroptosis susceptibility. Proteomic analysis identified 360 upregulated and 359 downregulated differentially expressed proteins between CuA and CuD groups under IH; these proteins were mainly enriched in citrate cycle, oxidative phosphorylation, fatty acid metabolism, the peroxisome proliferator-activated receptor (PPAR)α pathway, and ferroptosis. In IH exposure, CuD significantly upregulated the ferroptosis-promoting factor arachidonyl-CoA synthetase long chain family member (ACSL)4. ACSL4 knockdown markedly eliminated CuD-induced ferroptosis and lipid accumulation in IH exposure. In conculsion, IH can lead to reduced hepatic copper reserves and secondary iron deposition, thereby inducing ferroptosis and subsequent MAFLD progression. Insufficient dietary copper may worsen IH-associated MAFLD.


Sujet(s)
Cuivre , Ferroptose , Hypoxie , Souris de lignée C57BL , Animaux , Cuivre/métabolisme , Cuivre/déficit , Mâle , Souris , Hypoxie/métabolisme , Humains , Cellules HepG2 , Foie/métabolisme , Foie/anatomopathologie , Stress oxydatif , Métabolisme lipidique , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Stéatose hépatique/étiologie , Fer/métabolisme , Coenzyme A ligases/métabolisme , Coenzyme A ligases/génétique , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique
13.
Nutrients ; 16(13)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38999756

RÉSUMÉ

Celiac disease (CD) is a chronic autoimmune disorder triggered by the ingestion of gluten-containing food by genetically predisposed individuals. Hence, treatment of CD consists of permanent avoidance of wheat, rye, barley, and other gluten-containing foods. Lifelong adherence to a gluten-free diet (GFD) improves the symptoms of CD, but recent evidence suggests it is also associated with a higher risk for hepatic steatosis and the coexistence or emergence of other cardiometabolic risk factors. Moreover, a higher risk for liver steatosis is also reported by some authors as a potential extraintestinal complication of the CD itself. Recent nomenclature changes designate the association between hepatic steatosis and at least one of five cardiometabolic risk factors as metabolic dysfunction-associated steatotic liver disease (MASLD). An extended network of potentially causative factors underlying the association between MAFLD and CD, before and after dietary therapy is implemented, was recently described. The individualized treatment of these patients is less supported by evidence, with most of the current recommendations relying on empiric clinical judgment. This review focuses on the causative associations between CD and hepatic injury, either as an extraintestinal manifestation of CD or a side effect of GFD, also referring to potential therapeutic strategies for these individuals.


Sujet(s)
Maladie coeliaque , Régime sans gluten , Humains , Maladie coeliaque/diétothérapie , Maladie coeliaque/complications , Stéatose hépatique/diétothérapie , Stéatose hépatique/étiologie , Facteurs de risque , Maladies métaboliques/diétothérapie , Maladies métaboliques/étiologie
14.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(3): 653-661, 2024 May 20.
Article de Chinois | MEDLINE | ID: mdl-38948274

RÉSUMÉ

Objective: Non-alcoholic fatty liver disease (NAFLD) and alcohol-associated fatty liver disease (ALD) are the most common chronic liver diseases. Hepatic steatosis is an early histological subtype of both NAFLD and ALD. Excessive alcohol consumption is widely known to lead to hepatic steatosis and subsequent liver damage. However, reported findings concerning the association between moderate alcohol consumption and hepatic steatosis remain inconsistent. Notably, alcohol consumption as a modifiable lifestyle behavior is likely to change over time, but most previous studies covered alcohol intake only once at baseline. These inconsistent findings from existing studies do not inform decision-making concerning policies and clinical guidelines, which are of greater interest to health policymakers and clinician-scientists. Additionally, recommendations on the types of alcoholic beverages are not available. Usually, assessing the effects of two or more hypothetical alcohol consumption interventions on hepatic steatosis provides answers to questions concerning the population risk of hepatic steatosis if everyone changes from heavy drinking to abstinence, or if everyone keeps on drinking moderately, or if everyone of the drinking population switches from red wine to beer? Thus, we simulated a target trial to estimate the effects of several hypothetical interventions, including changes in the amount of alcohol consumption or the types of alcoholic beverages consumed, on hepatic steatosis using longitudinal data, to inform decisions about alcohol-related policymaking and clinical care. Methods: This longitudinal study included 12687 participants from the UK Biobank (UKB), all of whom participated in both baseline and repeat surveys. We excluded participants with missing data related to components of alcohol consumption and fatty liver index (FLI) in the baseline and the repeat surveys, as well as those who had reported liver diseases or cancer at the baseline survey. We used FLI as an outcome indicator and divided the participants into non-, moderate, and heavy drinkers. The surrogate marker FLI has been endorsed by many international organizations' guidelines, such as the European Association for the Study of the Liver. The calculation of FLI was based on laboratory and anthropometric data, including triglyceride, gamma-glutamyl transferase, body mass index, and waist circumference. Participants responded to questions about the types of alcoholic beverages, which were defined in 5 categories, including red wine, white wine/fortified wine/champagne, beer or cider, spirits, and mixed liqueurs, along with the average weekly or monthly amounts of alcohol consumed. Alcohol consumption was defined as pure alcohol consumed per week and was calculated according to the amount of alcoholic beverages consumed per week and the average ethanol content by volume in each alcoholic beverage. Participants were categorized as non-drinkers, moderate drinkers, and heavy drinkers according to the amount of their alcohol consumption. Moderate drinking was defined as consuming no more than 210 g of alcohol per week for men and 140 g of alcohol per week for women. We defined the following hypothetical interventions for the amount of alcohol consumed: sustaining a certain level of alcohol consumption from baseline to the repeat survey (e.g., none to none, moderate to moderate, heavy to heavy) and changing from one alcohol consumption level to another (e.g., none to moderate, moderate to heavy). The hypothetical interventions for the types of alcoholic beverages were defined in a similar way to those for the amount of alcohol consumed (e.g., red wine to red wine, red wine to beer/cider). We applied the parametric g-formula to estimate the effect of each hypothetical alcohol consumption intervention on the FLI. To implement the parametric g-formula, we first modeled the probability of time-varying confounders and FLI conditional on covariates. We then used these conditional probabilities to estimate the FLI value if the alcohol consumption level of each participant was under a specific hypothetical intervention. The confidence interval was obtained by 200 bootstrap samples. Results: For the alcohol consumption from baseline to the repeat surveys, 6.65% of the participants were sustained non-drinkers, 63.68% were sustained moderate drinkers, and 14.74% were sustained heavy drinkers, while 8.39% changed from heavy drinking to moderate drinking. Regarding the types of alcoholic beverages from baseline to the repeat surveys, 27.06% of the drinkers sustained their intake of red wine. Whatever the baseline alcohol consumption level, the hypothetical interventions for increasing alcohol consumption from the baseline alcohol consumption were associated with a higher FLI than that of the sustained baseline alcohol consumption level. When comparing sustained non-drinking with the hypothetical intervention of changing from non-drinking to moderate drinking, the mean ratio of FLI was 1.027 (95% confidence interval [CI]: 0.997-1.057). When comparing sustained non-drinking with the hypothetical intervention of changing from non-drinking to heavy drinking, the mean ratio of FLI was 1.075 (95% CI: 1.042-1.108). When comparing sustained heavy drinking with the hypothetical intervention of changing from heavy drinking to moderate drinking, the mean ratio of FLI was 0.953 (95% CI: 0.938-0.968). The hypothetical intervention of changing to red wine in the UKB was associated with lower FLI levels, compared with sustained consumption of other types of alcoholic beverages. For example, when comparing sustaining spirits with the hypothetical intervention of changing from spirits to red wine, the mean ratio of FLI was 0.981 (95% CI: 0.948-1.014). Conclusions: Regardless of the current level of alcohol consumption, interventions that increase alcohol consumption could raise the risk of hepatic steatosis in Western populations. The findings of this study could inform the formulation of future practice guidelines and health policies. If quitting drinking is challenging, red wine may be a better option than other types of alcoholic beverages in Western populations.


Sujet(s)
Consommation d'alcool , Stéatose hépatique non alcoolique , Humains , Consommation d'alcool/effets indésirables , Consommation d'alcool/épidémiologie , Études longitudinales , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/épidémiologie , Mâle , Femelle , Boissons alcooliques/effets indésirables , Stéatose hépatique alcoolique/étiologie , Adulte d'âge moyen , Stéatose hépatique/étiologie , Études de cohortes
15.
BMC Endocr Disord ; 24(1): 126, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39054440

RÉSUMÉ

BACKGROUND: Metabolic syndrome (MetS) as a multifactorial disorder is associated with non-communicable diseases. The dietary approaches to stop hypertension (DASH) diet is a healthy dietary pattern. We investigated the effect of the DASH diet on fatty liver and cardiovascular risk factors in subjects with MetS. METHODS: 60 Subjects with MetS were assigned into the intervention group (DASH diet) or the control group (a healthy diet). Fatty liver index (FLI), hepatic steatosis index (HSI), waist circumference (WC), weight, body mass index (BMI), triglyceride (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-c) and high-density lipoprotein cholesterol (HDL-c) were evaluated at the beginning and after intervention. Equations of fatty liver indices such as FLI and HSI are based on liver enzymes, anthropometric variables, sex and having diabetes. RESULTS: 30 subjects in the intervention group and 29 subjects in the control group completed the study. We found a significant reduction in the intervention group compared to the control group in FLI (-13.06 ± 10.03 vs. -2.90 ± 6.82;P < 0.001), HSI (-2.72 ± 2.59 vs. -0.81 ± 3.80;P = 0.02), WC (-6.02 ± 4.24 vs. -2.24 ± 4.28;P = 0.001), weight (-3.39 ± 2.53 vs. -1.51 ± 2.72;P = 0.008), BMI (-1.25 ± 0.93 vs. -0.56 ± 1.01;P = 0.008), DBP (-5.16 ± 3.92 vs. -1.50 ± 7.04;P = 0.01), SBP (-6.97 ± 8.21 vs. -1.36 ± 6.83;P = 0.006), TG (-18.50 ± 14.32 vs. 0.60 ± 23.81;P < 0.001), TC (-16.10 ± 17.94 vs. -5.07 ± 23.62;P = 0.04) and LDL-c (-13.50 ± 9.58 vs. -4.90 ± 18.28;P = 0.02). These results remained significant after adjusting for confounding factors, except for TC (P = 0.25). CONCLUSIONS: The DASH diet was more effective than the control diet in managing fatty liver and cardiovascular risk factors. TRIAL REGISTRATION: The trial was registered on 21 October 2022 at Iranian Registry of Clinical Trials (IRCT20180201038585N12, URL: https://irct.behdasht.gov.ir/trial/66161 ).


Sujet(s)
Maladies cardiovasculaires , Régime DASH , Facteurs de risque de maladie cardiaque , Syndrome métabolique X , Humains , Mâle , Syndrome métabolique X/diétothérapie , Syndrome métabolique X/complications , Femelle , Régime DASH/méthodes , Adulte d'âge moyen , Adulte , Maladies cardiovasculaires/prévention et contrôle , Maladies cardiovasculaires/étiologie , Stéatose hépatique/diétothérapie , Stéatose hépatique/étiologie , Facteurs de risque , Études de suivi , Hypertension artérielle/diétothérapie , Hypertension artérielle/complications
16.
Sci Adv ; 10(30): eado3141, 2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39047111

RÉSUMÉ

Metabolic dysfunction-associated steatohepatitis (MASH) is regulated by complex interplay between the macrophages and surrounding cells in the liver. Here, we show that Atf3 regulates glucose-fatty acid cycle in macrophages attenuates hepatocyte steatosis, and fibrogenesis in hepatic stellate cells (HSCs). Overexpression of Atf3 in macrophages protects against the development of MASH in Western diet-fed mice, whereas Atf3 ablation has the opposite effect. Mechanistically, Atf3 improves the reduction of fatty acid oxidation induced by glucose via forkhead box O1 (FoxO1) and Cd36. Atf3 inhibits FoxO1 activity via blocking Hdac1-mediated FoxO1 deacetylation at K242, K245, and K262 and increases Zdhhc4/5-mediated CD36 palmitoylation at C3, C7, C464, and C466; furthermore, macrophage Atf3 decreases hepatocytes lipogenesis and HSCs activation via retinol binding protein 4 (Rbp4). Anti-Rbp4 can prevent MASH progression that is induced by Atf3 deficiency in macrophages. This study identifies Atf3 as a regulator of glucose-fatty acid cycle. Targeting macrophage Atf3 or Rbp4 may be a plausible therapeutic strategy for MASH.


Sujet(s)
Facteur de transcription ATF-3 , Macrophages , Animaux , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Souris , Macrophages/métabolisme , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Stéatose hépatique/étiologie , Cellules étoilées du foie/métabolisme , Acides gras/métabolisme , Glucose/métabolisme , Foie/métabolisme , Foie/anatomopathologie , Hépatocytes/métabolisme , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Lipogenèse , Humains , Protéine O1 à motif en tête de fourche/métabolisme , Protéine O1 à motif en tête de fourche/génétique , Reprogrammation cellulaire , Souris de lignée C57BL ,
17.
Int J Mol Sci ; 25(14)2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39062970

RÉSUMÉ

Diabetes mellitus (DM) significantly impacts renal and hepatic function, necessitating comprehensive understanding and management strategies. Renal involvement, namely diabetic kidney disease (DKD), presents a global challenge, with increasing prevalence paralleling DM rates. Lifestyle modifications and pharmacotherapy targeting hypertension and glycemic control have pivotal roles in DKD management. Concurrently, hepatic involvement in DM, characterized by metabolic dysfunction-associated steatotic liver disease (MASLD), presents a bidirectional relationship. DM exacerbates MASLD progression, while MASLD predisposes to DM development and worsens glycemic control. Screening for MASLD in DM patients is of high importance, utilizing non-invasive methods like ultrasound and fibrosis scores. Lifestyle modifications, such as weight loss and a Mediterranean diet, mitigate MASLD progression. Promising pharmacotherapies, like SGLT2 inhibitors and GLP-1 agonists, demonstrate efficacy in both DM and MASLD management. Special populations, such as diabetic individuals undergoing hemodialysis or kidney transplant recipients, demand special care due to unique clinical features. Similarly, DM exacerbates complications in MASLD patients, elevating the risks of hepatic decompensation and hepatocellular carcinoma. Recognizing the interconnectedness of DM, renal, and hepatic diseases underscores the need for multidisciplinary approaches for optimal patient outcomes. The present review aims to present the main characteristics and crucial points not to be overlooked regarding the renal and hepatic involvement in DM patients focusing on the inter-relationships between the renal and the hepatic involvements.


Sujet(s)
Diabète de type 2 , Néphropathies diabétiques , Humains , Diabète de type 2/complications , Diabète de type 2/métabolisme , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/thérapie , Néphropathies diabétiques/étiologie , Stéatose hépatique/thérapie , Stéatose hépatique/étiologie , Stéatose hépatique/métabolisme , Prise en charge de la maladie , Foie/métabolisme , Foie/anatomopathologie , Hypoglycémiants/usage thérapeutique
18.
Nutrients ; 16(12)2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38931165

RÉSUMÉ

Iron deficiency is the number one nutritional problem worldwide. Iron uptake is regulated at the intestine and is highly influenced by the gut microbiome. Blood from the intestines drains directly into the liver, informing iron status and gut microbiota status. Changes in either iron or the microbiome are tightly correlated with the development of metabolic dysfunction-associated steatotic liver disease (MASLD). To investigate the underlying mechanisms of the development of MASLD that connect altered iron metabolism and gut microbiota, we compared specific pathogen free (SPF) or germ-free (GF) mice, fed a normal or low-iron diet. SPF mice on a low-iron diet showed reduced serum triglycerides and MASLD. In contrast, GF low-iron diet-fed mice showed increased serum triglycerides and did not develop hepatic steatosis. SPF mice showed significant changes in liver lipid metabolism and increased insulin resistance that was dependent upon the presence of the gut microbiota. We report that total body loss of mitochondrial iron importer Mitoferrin2 (Mfrn2-/-) exacerbated the development of MASLD on a low-iron diet with significant lipid metabolism alterations. Our study demonstrates a clear contribution of the gut microbiome, dietary iron, and Mfrn2 in the development of MASLD and metabolic syndrome.


Sujet(s)
Microbiome gastro-intestinal , Foie , Animaux , Femelle , Mâle , Souris , Stéatose hépatique/étiologie , Insulinorésistance , Fer/métabolisme , Carences en fer , Fer alimentaire/administration et posologie , Métabolisme lipidique , Foie/métabolisme , Souris de lignée C57BL , Souris knockout , Mitochondries/métabolisme , Protéines mitochondriales/métabolisme , Triglycéride/sang
19.
In Vivo ; 38(4): 1677-1689, 2024.
Article de Anglais | MEDLINE | ID: mdl-38936893

RÉSUMÉ

BACKGROUND/AIM: Depression is associated with metabolic disorders, including non-alcoholic fatty liver disease (NAFLD). However, the mechanisms underlying the interaction between them are still poorly known. MATERIALS AND METHODS: In this study, mice on a choline deficiency, L-amino acid-defined, high-fat diet (CDAHFD) developing steatosis were challenged with chronic restraint stress (CRS), a protocol widely used to induce depression. The development of depression and steatosis was evaluated using histopathology analysis, ELISA, q-PCR and Western Blot. RESULTS: The contribution of the activated HPA axis to hepatic steatosis progress was fully established, which was validated using a hepatocyte model. Histopathological and biochemical analysis indicated that steatosis was exacerbated by CRS challenge, and behavioral tests indicated that the mice developed depression. Among the screened endocrinal pathways, the hypothalamic-pituitary-adrenal (HPA) axis was significantly activated and the synergistic effect of CDAHFD and CRS in activating the HPA axis was observed. In the hypothalamus, expression of corticotropin-releasing hormone (CRH) was increased by 86% and the protein levels of hypothalamic CRH were upregulated by 25% to 33% by CRS treatment. Plasma CRH levels were elevated by 45-56% and plasma adrenocorticotropic hormone (ACTH) levels were elevated by 29-58% by CRS treatment. In the liver, target genes of the HPA axis were activated, accompanied by disruption of the lipid metabolism and progression of steatohepatitis. The lipid metabolism in the Hepa1-6 cell line treated with endogenous corticosterone (CORT) was in accordance with the aforementioned in vivo responses. CONCLUSION: Depression aggravated hepatic steatosis in CDAHFD-fed mice by activating the HPA axis. The risk of NAFLD development should be fully considered in depressive patients and improvement of psychotic disorders could be an etiological treatment strategy for them.


Sujet(s)
Dépression , Modèles animaux de maladie humaine , Axe hypothalamohypophysaire , Stéatose hépatique non alcoolique , Axe hypophyso-surrénalien , Animaux , Axe hypothalamohypophysaire/métabolisme , Axe hypophyso-surrénalien/métabolisme , Souris , Dépression/métabolisme , Dépression/étiologie , Dépression/génétique , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/étiologie , Mâle , Souris de lignée C57BL , Corticolibérine/métabolisme , Corticolibérine/génétique , Alimentation riche en graisse/effets indésirables , Hormone corticotrope/sang , Foie/métabolisme , Foie/anatomopathologie , Stéatose hépatique/métabolisme , Stéatose hépatique/étiologie , Stéatose hépatique/anatomopathologie , Corticostérone/sang
20.
Sci Rep ; 14(1): 14621, 2024 06 25.
Article de Anglais | MEDLINE | ID: mdl-38918525

RÉSUMÉ

Gestational diabetes mellitus (GDM) is associated with increased postpartum risk for metabolic dysfunction-associated steatotic liver disease (MASLD). GDM-related MASLD predisposes to advanced liver disease, necessitating a better understanding of its development in GDM. This preclinical study evaluated the MASLD development in a lean GDM mouse model with impaired insulin secretion capacity. Lean GDM was induced by short-term 60% high-fat diet and low-dose streptozotocin injections (60 mg/kg for 3 days) before mating in C57BL/6N mice. The control dams received only high-fat diet or low-fat diet. Glucose homeostasis was assessed during pregnancy and postpartum, whereas MASLD was assessed on postpartum day 30 (PP30). GDM dams exhibited a transient hyperglycemic phenotype during pregnancy, with hyperglycaemia reappearing after lactation. Lower insulin levels and impaired glucose-induced insulin response were observed in GDM mice during pregnancy and postpartum. At PP30, GDM dams displayed higher hepatic triglyceride content compared controls, along with increased MAS (MASLD) activity scores, indicating lipid accumulation, inflammation, and cell turnover indices. Additionally, at PP30, GDM dams showed elevated plasma liver injury markers. Given the absence of obesity in this double-hit GDM model, the results clearly indicate that impaired insulin secretion driven pregnancy hyperglycaemia has a distinct contribution to the development of postpartum MASLD.


Sujet(s)
Diabète gestationnel , Modèles animaux de maladie humaine , Souris de lignée C57BL , Période du postpartum , Animaux , Diabète gestationnel/métabolisme , Grossesse , Femelle , Souris , Période du postpartum/métabolisme , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Stéatose hépatique/étiologie , Insuline/métabolisme , Insuline/sang , Alimentation riche en graisse/effets indésirables , Foie/métabolisme , Foie/anatomopathologie , Glycémie/métabolisme , Triglycéride/métabolisme , Triglycéride/sang
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE