Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.927
Filtrer
1.
Clin Transl Sci ; 17(6): e13760, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38847320

RÉSUMÉ

Metabolic dysfunction-associated steatohepatitis (MASH) is the severe form of non-alcoholic fatty liver disease which has a high potential to progress to cirrhosis and hepatocellular carcinoma, yet adequate effective therapies are lacking. Hypoadiponectinemia is causally involved in the pathogenesis of MASH. This study investigated the pharmacological effects of adiponectin replacement therapy with the adiponectin-derived peptide ALY688 (ALY688-SR) in a mouse model of MASH. Human induced pluripotent stem (iPS) cell-derived hepatocytes were used to test cytotoxicity and signaling of unmodified ALY688 in vitro. High-fat diet with low methionine and no added choline (CDAHF) was used to induce MASH and test the effects of ALY688-SR in vivo. Histological MASH activity score (NAS) and fibrosis score were determined to assess the effect of ALY688-SR. Transcriptional characterization of mice through RNA sequencing was performed to indicate potential molecular mechanisms involved. In cultured hepatocytes, ALY688 efficiently induced adiponectin-like signaling, including the AMP-activated protein kinase and p38 mitogen-activated protein kinase pathways, and did not elicit cytotoxicity. Administration of ALY688-SR in mice did not influence body weight but significantly ameliorated CDAHF-induced hepatic steatosis, inflammation, and fibrosis, therefore effectively preventing the development and progression of MASH. Mechanistically, ALY688-SR treatment markedly induced hepatic expression of genes involved in fatty acid oxidation, whereas it significantly suppressed the expression of pro-inflammatory and pro-fibrotic genes as demonstrated by transcriptomic analysis. ALY688-SR may represent an effective approach in MASH treatment. Its mode of action involves inhibition of hepatic steatosis, inflammation, and fibrosis, possibly via canonical adiponectin-mediated signaling.


Sujet(s)
Adiponectine , Modèles animaux de maladie humaine , Hépatocytes , Stéatose hépatique non alcoolique , Animaux , Adiponectine/métabolisme , Adiponectine/pharmacologie , Adiponectine/déficit , Souris , Humains , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/prévention et contrôle , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/étiologie , Mâle , Souris de lignée C57BL , Transduction du signal/effets des médicaments et des substances chimiques , Alimentation riche en graisse/effets indésirables , Erreurs innées du métabolisme/métabolisme , Erreurs innées du métabolisme/traitement médicamenteux , Erreurs innées du métabolisme/anatomopathologie , Maladies métaboliques/traitement médicamenteux , Maladies métaboliques/métabolisme , Maladies métaboliques/prévention et contrôle , Maladies métaboliques/étiologie , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Stéatose hépatique/prévention et contrôle , Stéatose hépatique/métabolisme , Stéatose hépatique/traitement médicamenteux , Stéatose hépatique/anatomopathologie
2.
J Obes ; 2024: 7204607, 2024.
Article de Anglais | MEDLINE | ID: mdl-38831961

RÉSUMÉ

Obesity is a complex chronic disease characterized by excess body fat (adipose) that is harmful to health and has been a major global health problem. It may be associated with several diseases, such as nonalcoholic fatty liver disease (NAFLD). Polyunsaturated fatty acids (PUFA) are lipid mediators that have anti-inflammatory characteristics and can be found in animals and plants, with capybara oil (CO) being a promising source. So, we intend to evaluate the hepatic pathophysiological alterations in C57Bl/6 mice with NAFLD, caused by obesity, and the possible beneficial effects of OC in the treatment of this disease. Eighteen 3-month-old male C57Bl/6 mice received a control or high-fat diet for 18 weeks. From the 15th to the 18th week, the animals received treatment-through orogastric gavage-with placebo or free capybara oil (5 g/kg). Parameters inherent to body mass, glucose tolerance, evaluation of liver enzymes, percentage of hepatic steatosis, oxidative stress, the process of cell death with the apoptotic biomarkers (Bax, Bcl2, and Cytochrome C), and the ultrastructure of hepatocytes were analyzed. Even though the treatment with CO was not able to disassemble the effects on the physiological parameters, it proved to be beneficial in reversing the morphological and ultrastructural damage present in the hepatocytes. Thus, demonstrating that CO has beneficial effects in reducing steatosis and the apoptotic pathway, it is a promising treatment for NAFLD.


Sujet(s)
Apoptose , Foie , Stéatose hépatique non alcoolique , Huiles , Rodentia , Souris de lignée C57BL , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/thérapie , Mâle , Animaux , Souris , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/anatomopathologie , Hépatocytes/ultrastructure , Huiles/pharmacologie , Huiles/usage thérapeutique , Obésité/complications , Apoptose/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Foie/ultrastructure , Oxidoreductases/métabolisme , Activation enzymatique/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques
3.
J Appl Microbiol ; 135(6)2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38830802

RÉSUMÉ

AIMS: The incidence of nonalcoholic fatty liver disease (NAFLD) is increasing annually, leading to substantial medical and health burdens. Numerous studies have demonstrated the potential effectiveness of intestinal probiotics as a treatment strategy for NAFLD. Therefore, the objective of this study is to identify a probiotic for the treatment of NAFLD. METHODS AND RESULTS: In this study, blood and fecal samples were collected from 41 healthy volunteers and 44 patients diagnosed with NAFLD. Analysis of the 16S rDNA sequencing data and quantitative real-time PCR (RT-qPCR) revealed a significant reduction in the abundance of Coprococcus in NAFLD patients. Subsequent animal experiments demonstrated that Coprococcus was able to effectively reverse liver lipid accumulation, inflammation, and fibrosis induced by a high-fat diet (HFD) in mice. CONCLUSIONS: This study provides the first in vivo evidence that Coprococcus is a beneficial bacterium capable of preventing NAFLD and has the same probiotic effect in mice as Lactobacillus GG (LGG), a positive control. Therefore, Coprococcus has the potential to serve as a probiotic for the prevention and treatment of NAFLD in humans.


Sujet(s)
Alimentation riche en graisse , Stéatose hépatique non alcoolique , Probiotiques , Animaux , Stéatose hépatique non alcoolique/prévention et contrôle , Stéatose hépatique non alcoolique/microbiologie , Stéatose hépatique non alcoolique/étiologie , Alimentation riche en graisse/effets indésirables , Probiotiques/pharmacologie , Probiotiques/usage thérapeutique , Souris , Humains , Mâle , Souris de lignée C57BL , Fèces/microbiologie , Fèces/composition chimique , Adulte , Femelle , Foie/métabolisme , Microbiome gastro-intestinal , Adulte d'âge moyen , Modèles animaux de maladie humaine
4.
Zhonghua Gan Zang Bing Za Zhi ; 32(5): 418-434, 2024 May 20.
Article de Chinois | MEDLINE | ID: mdl-38858192

RÉSUMÉ

The Chinese Society of Hepatology of the Chinese Medical Association invited relevant experts to revise and update the Guideline of Prevention and Treatment of Nonalcoholic Fatty Liver Disease (2018Version) and renamed it as (Version 2024) Guideline for the Prevention and Treatment of Metabolic Dysfunction-associated (non-alcoholic) Fatty Liver Disease. Herein, the guiding recommendations on clinical issues such as screening and monitoring, diagnosis and evaluation, treatment and follow-up of metabolic dysfunction-associated fatty liver disease are put forward.


Sujet(s)
Stéatose hépatique non alcoolique , Stéatose hépatique non alcoolique/thérapie , Stéatose hépatique non alcoolique/prévention et contrôle , Stéatose hépatique non alcoolique/diagnostic , Stéatose hépatique non alcoolique/étiologie , Humains , Maladies métaboliques/prévention et contrôle , Maladies métaboliques/thérapie , Maladies métaboliques/étiologie , Facteurs de risque , Chine
5.
Commun Biol ; 7(1): 681, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38831027

RÉSUMÉ

Metabolic dysfunction-associated steatohepatitis (MASH), previously called non-alcoholic steatohepatitis (NASH), is a growing concern worldwide, with liver fibrosis being a critical determinant of its prognosis. Monocyte-derived macrophages have been implicated in MASH-associated liver fibrosis, yet their precise roles and the underlying differentiation mechanisms remain elusive. In this study, we unveil a key orchestrator of this process: long chain saturated fatty acid-Egr2 pathway. Our findings identify the transcription factor Egr2 as the driving force behind monocyte differentiation into hepatic lipid-associated macrophages (hLAMs) within MASH liver. Notably, Egr2-deficiency reroutes monocyte differentiation towards a macrophage subset resembling resident Kupffer cells, hampering hLAM formation. This shift has a profound impact, suppressing the transition from benign steatosis to liver fibrosis, demonstrating the critical pro-fibrotic role played by hLAMs in MASH pathogenesis. Long-chain saturated fatty acids that accumulate in MASH liver emerge as potent inducers of Egr2 expression in macrophages, a process counteracted by unsaturated fatty acids. Furthermore, oral oleic acid administration effectively reduces hLAMs in MASH mice. In conclusion, our work not only elucidates the intricate interplay between saturated fatty acids, Egr2, and monocyte-derived macrophages but also highlights the therapeutic promise of targeting the saturated fatty acid-Egr2 axis in monocytes for MASH management.


Sujet(s)
Différenciation cellulaire , Facteur de transcription EGR-2 , Cirrhose du foie , Macrophages , Monocytes , Stéatose hépatique non alcoolique , Animaux , Facteur de transcription EGR-2/métabolisme , Facteur de transcription EGR-2/génétique , Souris , Monocytes/métabolisme , Macrophages/métabolisme , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/étiologie , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Cirrhose du foie/étiologie , Cirrhose du foie/génétique , Souris de lignée C57BL , Mâle , Modèles animaux de maladie humaine , Acides gras/métabolisme , Foie/métabolisme , Foie/anatomopathologie , Antigènes Ly
6.
World J Gastroenterol ; 30(20): 2633-2637, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38855152

RÉSUMÉ

This editorial builds on the article titled "Establishment and validation of an adherence prediction system for lifestyle interventions in non-alcoholic fatty liver disease" by Zeng et al. We carried out a critical examination of nonalcoholic fatty liver disease (NAFLD) pathogenesis and how lifestyle interventions could facilitate disease resolution, particularly highlighting that non-alcoholic steatohepatitis (NASH) is a severe form of NAFLD. Our discussion details that weight loss is a pivotal factor in disease outcomes: A 3%-5% reduction is enough for resolution in 50% of non-obese individuals, while a 7%-10% reduction achieves similar benefits in obese individuals, as demonstrated by magnetic resonance spectroscopy. Additionally, the editorial underscores that such lifestyle changes are instrumental not only in resolving NAFLD but also in reversing hepatic steatosis and inflammation. These insights, derived from the research, emphasize the critical role of personalized lifestyle modifications in halting the progression of NAFLD to NASH and even reversing fibrosis, thus offering a template for effective patient management.


Sujet(s)
Évolution de la maladie , Mode de vie , Stéatose hépatique non alcoolique , Perte de poids , Stéatose hépatique non alcoolique/thérapie , Stéatose hépatique non alcoolique/diagnostic , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/anatomopathologie , Humains , Obésité/diagnostic , Obésité/complications , Foie/anatomopathologie , Comportement de réduction des risques , Cirrhose du foie/thérapie , Cirrhose du foie/anatomopathologie , Résultat thérapeutique
7.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38892467

RÉSUMÉ

Nonalcoholic fatty liver disease (NAFLD) is one of the most common chronic liver diseases in the world, which begins with liver lipid accumulation and is associated with metabolic syndrome. Also, the name chosen to replace NAFLD was metabolic dysfunction-associated steatotic liver disease (MASLD). We performed focused drug screening and found that Cilostazol effectively ameliorated hepatic steatosis and might offer potential for NAFLD treatment. Our aim was to investigate the therapeutic effects of Cilostazol on the glycolipid metabolism and intestinal flora in NAFLD mice and explore the specific mechanism. In this study, 7-week-old male C57BL/6J mice were fed a high-fat diet (HFD) for 8 weeks to induce NAFLD, and then treated with intragastric administration for 12 weeks. The results showed that Cilostazol inhibited liver lipid de novo synthesis by regulating the AMPK-ACC1/SCD1 pathway and inhibited liver gluconeogenesis by the AMPK-PGC1α-G6P/PEPCK pathway. Cilostazol improved the intestinal flora diversity and intestinal microbial composition in the NAFLD mice, and specifically regulated Desulfovibrio and Akkermansia. In addition, Cilostazol increased the level of short-chain fatty acids in the NAFLD mice to a level similar to that in the blank Control group. Cilostazol reduces liver lipid accumulation in NAFLD mice by improving glucose and lipid metabolism disorders and intestinal dysfunction, thereby achieving the purpose of treating NAFLD.


Sujet(s)
Cilostazol , Microbiome gastro-intestinal , Métabolisme lipidique , Souris de lignée C57BL , Stéatose hépatique non alcoolique , Animaux , Cilostazol/pharmacologie , Cilostazol/usage thérapeutique , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/étiologie , Souris , Mâle , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Métabolisme lipidique/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Alimentation riche en graisse/effets indésirables , Maladies intestinales/traitement médicamenteux , Maladies intestinales/métabolisme , Modèles animaux de maladie humaine
8.
Molecules ; 29(11)2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38893506

RÉSUMÉ

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a significant hepatic condition that has gained worldwide attention. Kaempferol (Kae), renowned for its diverse biological activities, including anti-inflammatory, antioxidant, anti-aging, and cardio-protective properties, has emerged as a potential therapeutic candidate for non-alcoholic steatohepatitis (NASH). Despite its promising therapeutic potential, the precise underlying mechanism of Kae's beneficial effects in NASH remains unclear. Therefore, this study aims to clarify the mechanism by conducting comprehensive in vivo and in vitro experiments. RESULTS: In this study, a murine model of non-alcoholic steatohepatitis (NASH) was established by feeding C57BL/6 female mice a high-fat diet for 12 weeks. Kaempferol (Kae) was investigated for its ability to modulate systemic inflammatory responses and lipid metabolism in this model (20 mg/kg per day). Notably, Kae significantly reduced the expression of NLRP3-ASC/TMS1-Caspase 3, a crucial mediator of liver tissue inflammation. Additionally, in a HepG2 cell model induced with palmitic acid/oleic acid (PA/OA) to mimic NASH conditions, Kae demonstrated the capacity to decrease lipid droplet accumulation and downregulate the expression of NLRP3-ASC/TMS1-Caspase 3 (20 µM and the final concentration to 20 nM). These findings suggest that Kae may hold therapeutic potential in the treatment of NASH by targeting inflammatory and metabolic pathways. CONCLUSIONS: These findings suggest that kaempferol holds potential as a promising therapeutic intervention for ameliorating non-alcoholic fatty liver disease (NAFLD).


Sujet(s)
Caspase-3 , Kaempférols , Protéine-3 de la famille des NLR contenant un domaine pyrine , Granulocytes neutrophiles , Stéatose hépatique non alcoolique , Transduction du signal , Kaempférols/pharmacologie , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/anatomopathologie , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Souris , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Caspase-3/métabolisme , Femelle , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Granulocytes neutrophiles/métabolisme , Modèles animaux de maladie humaine , Souris de lignée C57BL , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Cellules HepG2 , Alimentation riche en graisse/effets indésirables
9.
Expert Rev Endocrinol Metab ; 19(4): 335-348, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38860684

RÉSUMÉ

INTRODUCTION: Obesity and metabolic-associated fatty liver disease (MAFLD) during pregnancy constitute significant problems for routine antenatal care, with increasing prevalence globally. Similar to obesity, MAFLD is associated with a higher risk for maternal complications (e.g. pre-eclampsia and gestational diabetes) and long-term adverse health outcomes for the offspring. However, MAFLD during pregnancy is often under-recognized, with limited management/treatment options. AREAS COVERED: PubMed/MEDLINE, EMBASE, and Scopus were searched based on a search strategy for obesity and/or MAFLD in pregnancy to identify relevant papers up to 2024. This review summarizes the pertinent evidence on the relationship between maternal obesity and MAFLD during pregnancy. Key mechanisms implicated in the underlying pathophysiology linking obesity and MAFLD during pregnancy (e.g. insulin resistance and dysregulated adipokine secretion) are highlighted. Moreover, a diagnostic approach for MAFLD diagnosis during pregnancy and its complications are presented. Finally, promising relevant areas for future research are covered. EXPERT OPINION: Research progress regarding maternal obesity, MAFLD, and their impact on maternal and fetal/offspring health is expected to improve the relevant diagnostic methods and lead to novel treatments. Thus, routine practice could apply more personalized management strategies, incorporating individualized algorithms with genetic and/or multi-biomarker profiling to guide prevention, early diagnosis, and treatment.


Sujet(s)
Obésité maternelle , Complications de la grossesse , Humains , Grossesse , Femelle , Obésité maternelle/complications , Obésité maternelle/métabolisme , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/étiologie
10.
Int J Mol Sci ; 25(11)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38891768

RÉSUMÉ

Gut-dysbiosis-induced lipopolysaccharides (LPS) translocation into systemic circulation has been suggested to be implicated in nonalcoholic fatty liver disease (NAFLD) pathogenesis. This study aimed to assess if oleuropein (OLE), a component of extra virgin olive oil, lowers high-fat-diet (HFD)-induced endotoxemia and, eventually, liver steatosis. An immunohistochemistry analysis of the intestine and liver was performed in (i) control mice (CTR; n = 15), (ii) high-fat-diet fed (HFD) mice (HFD; n = 16), and (iii) HFD mice treated with 6 µg/day of OLE for 30 days (HFD + OLE, n = 13). The HFD mice developed significant liver steatosis compared to the controls, an effect that was significantly reduced in the HFD + OLE-treated mice. The amount of hepatocyte LPS localization and the number of TLR4+ macrophages were higher in the HFD mice in the than controls and were lowered in the HFD + OLE-treated mice. The number of CD42b+ platelets was increased in the liver sinusoids of the HFD mice compared to the controls and decreased in the HFD + OLE-treated mice. Compared to the controls, the HFD-treated mice showed a high percentage of intestine PAS+ goblet cells, an increased length of intestinal crypts, LPS localization and TLR4+ expression, and occludin downregulation, an effect counteracted in the HFD + OLE-treated mice. The HFD-fed animals displayed increased systemic levels of LPS and zonulin, but they were reduced in the HFD + OLE-treated animals. It can be seen that OLE administration improves liver steatosis and inflammation in association with decreased LPS translocation into the systemic circulation, hepatocyte localization of LPS and TLR4 downregulation in HFD-induced mouse model of NAFLD.


Sujet(s)
Glucosides d'iridoïdes , Iridoïdes , Lipopolysaccharides , Stéatose hépatique non alcoolique , Huile d'olive , Récepteur de type Toll-4 , Animaux , Récepteur de type Toll-4/métabolisme , Glucosides d'iridoïdes/pharmacologie , Souris , Huile d'olive/pharmacologie , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/anatomopathologie , Mâle , Iridoïdes/pharmacologie , Régulation négative/effets des médicaments et des substances chimiques , Alimentation riche en graisse/effets indésirables , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Souris de lignée C57BL , Inflammation/métabolisme , Stéatose hépatique/métabolisme , Stéatose hépatique/traitement médicamenteux , Stéatose hépatique/étiologie , Stéatose hépatique/anatomopathologie
11.
Int J Mol Sci ; 25(11)2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38891828

RÉSUMÉ

The epidemiological burden of liver steatosis associated with metabolic diseases is continuously growing worldwide and in all age classes. This condition generates possible progression of liver damage (i.e., inflammation, fibrosis, cirrhosis, hepatocellular carcinoma) but also independently increases the risk of cardio-metabolic diseases and cancer. In recent years, the terminological evolution from "nonalcoholic fatty liver disease" (NAFLD) to "metabolic dysfunction-associated fatty liver disease" (MAFLD) and, finally, "metabolic dysfunction-associated steatotic liver disease" (MASLD) has been paralleled by increased knowledge of mechanisms linking local (i.e., hepatic) and systemic pathogenic pathways. As a consequence, the need for an appropriate classification of individual phenotypes has been oriented to the investigation of innovative therapeutic tools. Besides the well-known role for lifestyle change, a number of pharmacological approaches have been explored, ranging from antidiabetic drugs to agonists acting on the gut-liver axis and at a systemic level (mainly farnesoid X receptor (FXR) agonists, PPAR agonists, thyroid hormone receptor agonists), anti-fibrotic and anti-inflammatory agents. The intrinsically complex pathophysiological history of MASLD makes the selection of a single effective treatment a major challenge, so far. In this evolving scenario, the cooperation between different stakeholders (including subjects at risk, health professionals, and pharmaceutical industries) could significantly improve the management of disease and the implementation of primary and secondary prevention measures. The high healthcare burden associated with MASLD makes the search for new, effective, and safe drugs a major pressing need, together with an accurate characterization of individual phenotypes. Recent and promising advances indicate that we may soon enter the era of precise and personalized therapy for MASLD/MASH.


Sujet(s)
Stéatose hépatique non alcoolique , Humains , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/thérapie , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/complications , Stéatose hépatique non alcoolique/étiologie , Maladies métaboliques/métabolisme , Maladies métaboliques/étiologie , Stéatose hépatique/métabolisme , Stéatose hépatique/étiologie , Stéatose hépatique/thérapie , Stéatose hépatique/complications , Animaux
12.
Int J Mol Sci ; 25(11)2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38892011

RÉSUMÉ

Non-alcoholic fatty liver disease (NAFLD) is a clinicopathological syndrome characterized by diffuse hepatocellular steatosis due to fatty deposits in hepatocytes, excluding alcohol and other known liver injury factors. However, there are no specific drugs for the clinical treatment of NAFLD. Therefore, research on the pathogenesis of NAFLD at the cellular and molecular levels is a promising approach to finding therapeutic targets and developing targeted drugs for NAFLD. Pin1 is highly expressed during adipogenesis and contributes to adipose differentiation, but its specific mechanism of action in NAFLD is unclear. In this study, we investigated the role of Pin1 in promoting the development of NAFLD and its potential mechanisms in vitro and in vivo. First, Pin1 was verified in the NAFLD model in vitro using MCD diet-fed mice by Western Blot, RT-qPCR and immunohistochemistry (IHC) assays. In the in vitro study, we used the oleic acid (OA) stimulation-induced lipid accumulation model and examined the lipid accumulation in each group of cells by oil red O staining as well as BODIPY staining. The results showed that knockdown of Pin1 inhibited lipid accumulation in hepatocytes in an in vitro lipid accumulation model and improved lipid indices and liver injury levels. Moreover, in vivo, WT and Pin1-KO mice were fed a methionine-choline deficient (MCD) diet for 4 weeks to induce the NAFLD model. The effects of Pin1 on lipid accumulation, hepatic fibrosis, and oxidative stress were evaluated by biochemical analysis, glucose and insulin tolerance tests, histological analysis, IHC, RT-qPCR and Western blot assays. The results indicate that Pin1 knockdown significantly alleviated hepatic steatosis, fibrosis and inflammation in MCD-induced NAFLD mice, improved glucose tolerance and alleviated insulin resistance in mice. Further studies showed that the AMPK/ACC1 signalling pathway might take part in the process by which Pin1 regulates NAFLD, as evidenced by the inhibition of the AMPK/ACC1 pathway. In addition, immunofluorescence (IF), coimmunoprecipitation (Co-IP) and GST pull-down experiments also showed that Pin1 interacts directly with ACC1 and inhibits ACC1 phosphorylation levels. Our study suggests that Pin1 promotes NAFLD progression by inhibiting the activation of the AMPK/ACC1 signalling pathway, and it is possible that this effect is achieved by Pin1 interacting with ACC1 and inhibiting the phosphorylation of ACC1.


Sujet(s)
NIMA-interacting peptidylprolyl isomerase , Stéatose hépatique non alcoolique , Animaux , NIMA-interacting peptidylprolyl isomerase/métabolisme , NIMA-interacting peptidylprolyl isomerase/génétique , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/étiologie , Souris , Mâle , Souris knockout , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Humains , Métabolisme lipidique , Souris de lignée C57BL , Modèles animaux de maladie humaine , Liaison aux protéines , Acetyl-coA carboxylase
13.
Front Endocrinol (Lausanne) ; 15: 1371444, 2024.
Article de Anglais | MEDLINE | ID: mdl-38836220

RÉSUMÉ

Objective: Individuals with hypopituitarism (HPs) have an increased risk of developing non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH) due to growth hormone deficiency (GHD). We aimed to investigate the possible mechanisms underlying the relationship between GHD and NAFLD using proteomic and metabolomic insights. Methods: Serum metabolic alternations were assessed in male HPs using untargeted metabolomics. A rat model of HP was established through hypophysectomy, followed by recombinant human growth hormone (rhGH) intervention. The mechanisms underlying GHD-mediated NAFLD were elucidated through the application of label-free proteomics and phosphorylation proteomics. Results: Metabolomic analysis revealed that biomarkers of mitochondrial dysfunction and oxidative stress, such as alanine, lactate, and creatine, were significantly elevated in HPs compared to age-matched controls. In rats, hypophysectomy led to marked hepatic steatosis, lipid peroxidation, and reduced glutathione (GSH), which were subsequently modulated by rhGH replacement. Proteomic analysis identified cytochrome P450s, mitochondrial translation elongation, and PPARA activating genes as the major distinguishing pathways in hypophysectomized rats. The processes of fatty acid transport, synthesis, oxidation, and NADP metabolism were tightly described. An enhanced regulation of peroxisome ß-oxidation and ω-oxidation, together with a decreased NADPH regeneration, may exacerbate oxidative stress. Phosphoproteome data showed downregulation of JAK2-STAT5B and upregulation of mTOR signaling pathway. Conclusions: This study identified proteo-metabolomic signatures associated with the development of NAFLD in pituitary GHD. Evidence was found of oxidative stress imbalance resulting from abnormal fatty acid oxidation and NADPH regeneration, highlighting the role of GH deficiency in the development of NAFLD.


Sujet(s)
Hypopituitarisme , Métabolomique , Stéatose hépatique non alcoolique , Stress oxydatif , Protéomique , Animaux , Mâle , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/étiologie , Rats , Hypopituitarisme/métabolisme , Hypopituitarisme/étiologie , Rat Sprague-Dawley , Hormone de croissance humaine/déficit , Hormone de croissance humaine/métabolisme , Humains
14.
Nat Metab ; 6(6): 1178-1196, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38867022

RÉSUMÉ

Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease, encompasses steatosis and metabolic dysfunction-associated steatohepatitis (MASH), leading to cirrhosis and hepatocellular carcinoma. Preclinical MASLD research is mainly performed in rodents; however, the model that best recapitulates human disease is yet to be defined. We conducted a wide-ranging retrospective review (metabolic phenotype, liver histopathology, transcriptome benchmarked against humans) of murine models (mostly male) and ranked them using an unbiased MASLD 'human proximity score' to define their metabolic relevance and ability to induce MASH-fibrosis. Here, we show that Western diets align closely with human MASH; high cholesterol content, extended study duration and/or genetic manipulation of disease-promoting pathways are required to intensify liver damage and accelerate significant (F2+) fibrosis development. Choline-deficient models rapidly induce MASH-fibrosis while showing relatively poor translatability. Our ranking of commonly used MASLD models, based on their proximity to human MASLD, helps with the selection of appropriate in vivo models to accelerate preclinical research.


Sujet(s)
Modèles animaux de maladie humaine , Stéatose hépatique non alcoolique , Animaux , Humains , Souris , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/anatomopathologie , Mâle , Foie/métabolisme , Foie/anatomopathologie , Maladies métaboliques/métabolisme , Maladies métaboliques/étiologie , Régime occidental/effets indésirables , Études rétrospectives , Cirrhose du foie/métabolisme , Cirrhose du foie/étiologie
15.
Front Public Health ; 12: 1368483, 2024.
Article de Anglais | MEDLINE | ID: mdl-38746002

RÉSUMÉ

Background: The association between air pollution, lung function, gastroesophageal reflux disease, and Non-alcoholic fatty liver disease (NAFLD) remains inconclusive. Previous studies were not convincing due to confounding factors and reverse causality. We aim to investigate the causal relationship between air pollution, lung function, gastroesophageal reflux disease, and NAFLD using Mendelian randomization analysis. Methods: In this study, univariate Mendelian randomization analysis was conducted first. Subsequently, Steiger testing was performed to exclude the possibility of reverse association. Finally, significant risk factors identified from the univariate Mendelian analysis, as well as important factors affecting NAFLD from previous observational studies (type 2 diabetes and body mass index), were included in the multivariable Mendelian randomization analysis. Results: The results of the univariable Mendelian randomization analysis showed a positive correlation between particulate matter 2.5, gastroesophageal reflux disease, and NAFLD. There was a negative correlation between forced expiratory volume in 1 s, forced vital capacity, and NAFLD. The multivariable Mendelian randomization analysis indicated a direct causal relationship between gastroesophageal reflux disease (OR = 1.537, p = 0.011), type 2 diabetes (OR = 1.261, p < 0.001), and NAFLD. Conclusion: This Mendelian randomization study confirmed the causal relationships between air pollution, lung function, gastroesophageal reflux, and NAFLD. Furthermore, gastroesophageal reflux and type 2 diabetes were identified as independent risk factors for NAFLD, having a direct causal connection with the occurrence of NAFLD.


Sujet(s)
Pollution de l'air , Reflux gastro-oesophagien , Analyse de randomisation mendélienne , Stéatose hépatique non alcoolique , Humains , Reflux gastro-oesophagien/génétique , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/génétique , Pollution de l'air/effets indésirables , Facteurs de risque , Diabète de type 2/épidémiologie , Diabète de type 2/génétique , Tests de la fonction respiratoire , Matière particulaire/effets indésirables , Mâle , Femelle , Causalité
16.
J Oleo Sci ; 73(5): 695-708, 2024.
Article de Anglais | MEDLINE | ID: mdl-38692892

RÉSUMÉ

This study was to investigate the effects of Smilax China L. saponins (SCS) on non-alcoholic fatty liver disease (NAFLD). Rats were fed a high-fat diet (HFD) for 8 weeks to induce NAFLD, followed by SCS treatment for 8 weeks. The effect of SCS on liver injury was observed by H&E staining and the regulative mechanism of SCS on lipid formation was exposed by detecting Oil red O, insulin resistance (IR), and fatty acids synthesis (FAS). Furthermore, transcriptomics and metabolomics were performed to analyze the potential targets. The experimental results indicated that SCS exerted a positive curative effect in alleviating HFD-induced overweight, hepatic injury, steatosis, and lipid formation and accumulation in rats, and the preliminary mechanism studies showed that SCS could alleviate IR, inhibit FAS expression, and reduce Acetyl-CoA levels. Besides, the integrative analysis of transcriptomics and metabolomics exposed the targets of SCS to regulate lipid production likely being the sphingolipid metabolism and glycerophospholipid metabolism pathways. This study demonstrates that SCS significantly ameliorates lipid metabolic disturbance in rats with NAFLD by relieving insulin resistance, inhibiting the FAS enzymes, and regulating the sphingolipid and glycerophospholipid metabolism pathways.


Sujet(s)
Alimentation riche en graisse , Insulinorésistance , Métabolisme lipidique , Métabolomique , Stéatose hépatique non alcoolique , Saponines , Smilax , Transcriptome , Animaux , Smilax/composition chimique , Saponines/pharmacologie , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/génétique , Mâle , Métabolomique/méthodes , Alimentation riche en graisse/effets indésirables , Transcriptome/effets des médicaments et des substances chimiques , Métabolisme lipidique/effets des médicaments et des substances chimiques , Rats , Rat Sprague-Dawley , Sphingolipides/métabolisme , Glycérophospholipides/métabolisme , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine
17.
Nutrients ; 16(9)2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38732634

RÉSUMÉ

Nonalcoholic fatty liver disease (NAFLD) has emerged as the most prevalent pediatric liver disorder, primarily attributed to dietary shifts in recent years. NAFLD is characterized by the accumulation of lipid species in hepatocytes, leading to liver inflammation that can progress to steatohepatitis, fibrosis, and cirrhosis. Risk factors contributing to NAFLD encompass genetic variations and metabolic disorders such as obesity, diabetes, and insulin resistance. Moreover, transgenerational influences, resulting in an imbalance of gut microbial composition, epigenetic modifications, and dysregulated hepatic immune responses in offspring, play a pivotal role in pediatric NAFLD development. Maternal nutrition shapes the profile of microbiota-derived metabolites in offspring, exerting significant influence on immune system regulation and the development of metabolic syndrome in offspring. In this review, we summarize recent evidence elucidating the intricate interplay between gut microbiota, epigenetics, and immunity in fetuses exposed to maternal nutrition, and its impact on the onset of NAFLD in offspring. Furthermore, potential therapeutic strategies targeting this network are also discussed.


Sujet(s)
Épigenèse génétique , Microbiome gastro-intestinal , Phénomènes physiologiques nutritionnels maternels , Stéatose hépatique non alcoolique , Effets différés de l'exposition prénatale à des facteurs de risque , Stéatose hépatique non alcoolique/immunologie , Stéatose hépatique non alcoolique/microbiologie , Stéatose hépatique non alcoolique/étiologie , Humains , Femelle , Grossesse , Animaux , Facteurs de risque
18.
Clin Nutr ; 43(6): 1503-1515, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38729079

RÉSUMÉ

BACKGROUND & AIMS: Nonalcoholic fatty liver disease (NAFLD) is related to muscle loss, but the precise mechanism underlying this association remains unclear. The aim of the present study was thus to determine the influence of maternal fatty liver and dietary choline deficiency during pregnancy and/or lactation periods on the skeletal muscle gene expression profile among 24-day-old male rat offspring. METHODS: Histological examination of skeletal muscle tissue specimens obtained from offspring of dams suffering from fatty liver, provided with proper choline intake during pregnancy and lactation (NN), fed a choline-deficient diet during both periods (DD), deprived of choline only during pregnancy (DN), or only during lactation (ND), was performed. The global transcriptome pattern was assessed using a microarray approach (Affymetrix® Rat Gene 2.1 ST Array Strip). The relative expression of selected genes was validated by real-time PCR (qPCR). RESULTS: Morphological differences in fat accumulation in skeletal muscle related to choline supply were observed. The global gene expression profile was consistent with abnormal morphological changes. Mettl21c gene was overexpressed in all choline-deficient groups compared to the NN group, while two genes, Cdkn1a and S100a4, were downregulated. Processes of protein biosynthesis were upregulated, and processes related to cell proliferation and lipid metabolism were inhibited in DD, DN, and ND groups compared to the NN group. CONCLUSIONS: Prenatal and early postnatal exposure to fatty liver and dietary choline deficiency leads to changes in the transcriptome profile in skeletal muscle of 24-day old male rat offspring and is associated with muscle damage, but the mechanism of it seems to be different at different developmental stages of life. Adequate choline intake during pregnancy and lactation can prevent severe muscle disturbance in the progeny of females suffering from fatty liver.


Sujet(s)
Carence en choline , Choline , Lactation , Muscles squelettiques , Effets différés de l'exposition prénatale à des facteurs de risque , Transcriptome , Animaux , Femelle , Grossesse , Muscles squelettiques/métabolisme , Mâle , Rats , Choline/administration et posologie , Phénomènes physiologiques nutritionnels maternels , Rat Wistar , Régime alimentaire , Stéatose hépatique non alcoolique/génétique , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/étiologie
19.
Commun Biol ; 7(1): 594, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38760406

RÉSUMÉ

Non-alcoholic fatty liver disease (NAFLD) is a chronic disease caused by hepatic steatosis. Adenosine deaminases acting on RNA (ADARs) catalyze adenosine to inosine RNA editing. However, the functional role of ADAR2 in NAFLD is unclear. ADAR2+/+/GluR-BR/R mice (wild type, WT) and ADAR2-/-/GluR-BR/R mice (ADAR2 KO) mice are fed with standard chow or high-fat diet (HFD) for 12 weeks. ADAR2 KO mice exhibit protection against HFD-induced glucose intolerance, insulin resistance, and dyslipidemia. Moreover, ADAR2 KO mice display reduced liver lipid droplets in concert with decreased hepatic TG content, improved hepatic insulin signaling, better pyruvate tolerance, and increased glycogen synthesis. Mechanistically, ADAR2 KO effectively mitigates excessive lipid production via AMPK/Sirt1 pathway. ADAR2 KO inhibits hepatic gluconeogenesis via the AMPK/CREB pathway and promotes glycogen synthesis by activating the AMPK/GSK3ß pathway. These results provide evidence that ADAR2 KO protects against NAFLD progression through the activation of AMPK signaling pathways.


Sujet(s)
Adenosine deaminase , Alimentation riche en graisse , Souris knockout , Stéatose hépatique non alcoolique , Protéines de liaison à l'ARN , Transduction du signal , Animaux , Adenosine deaminase/métabolisme , Adenosine deaminase/génétique , Adenosine deaminase/déficit , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Souris , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/génétique , Stéatose hépatique non alcoolique/étiologie , Alimentation riche en graisse/effets indésirables , Mâle , AMP-Activated Protein Kinases/métabolisme , AMP-Activated Protein Kinases/génétique , Insulinorésistance , Souris obèse , Obésité/métabolisme , Obésité/génétique , Souris de lignée C57BL , Foie/métabolisme
20.
Front Endocrinol (Lausanne) ; 15: 1282231, 2024.
Article de Anglais | MEDLINE | ID: mdl-38756999

RÉSUMÉ

Introduction: Cigarettes containing nicotine (Nic) are a risk factor for the development of cardiovascular and metabolic diseases. We reported that Nic delivered via injections or e-cigarette vapor led to hepatic steatosis in mice fed with a high-fat diet. High-fructose corn syrup (HFCS) is the main sweetener in sugar-sweetened beverages (SSBs) in the US. Increased consumption of SSBs with HFCS is associated with increased risks of non-alcoholic fatty liver disease (NAFLD). Nicotinamide riboside (NR) increases mitochondrial nicotinamide adenine dinucleotide (NAD+) and protects mice against hepatic steatosis. This study evaluated if Nic plus Coca-Cola™ (Coke) with HFCS can cause hepatic steatosis and that can be protected by NR. Methods: C57BL/6J mice received twice daily intraperitoneal (IP) injections of Nic or saline and were given Coke (HFCS), or Coke with sugar, and NR supplementation for 10 weeks. Results: Our results show that Nic+Coke caused increased caloric intake and induced hepatic steatosis, and the addition of NR prevented these changes. Western blot analysis showed lipogenesis markers were activated (increased cleavage of the sterol regulatory element-binding protein 1 [SREBP1c] and reduction of phospho-Acetyl-CoA Carboxylase [p-ACC]) in the Nic+Coke compared to the Sal+Water group. The hepatic detrimental effects of Nic+Coke were mediated by decreased NAD+ signaling, increased oxidative stress, and mitochondrial damage. NR reduced oxidative stress and prevented mitochondrial damage by restoring protein levels of Sirtuin1 (Sirt1) and peroxisome proliferator-activated receptor coactivator 1-alpha (PGC1) signaling. Conclusion: We conclude that Nic+Coke has an additive effect on producing hepatic steatosis, and NR is protective. This study suggests concern for the development of NAFLD in subjects who consume nicotine and drink SSBs with HFCS.


Sujet(s)
Souris de lignée C57BL , Nicotinamide , Nicotine , Composés de pyridinium , Animaux , Composés de pyridinium/pharmacologie , Souris , Nicotinamide/analogues et dérivés , Nicotinamide/pharmacologie , Mâle , Stéatose hépatique non alcoolique/prévention et contrôle , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique/prévention et contrôle , Stéatose hépatique/métabolisme , Stéatose hépatique/induit chimiquement , Sirop de maïs à haute teneur en fructose/effets indésirables , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...