Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 9.482
Filtrer
1.
Zhongguo Zhong Yao Za Zhi ; 49(14): 3804-3817, 2024 Jul.
Article de Chinois | MEDLINE | ID: mdl-39099354

RÉSUMÉ

The chemical composition of Ganoderma lucidum ethanol extracts was systematically analyzed and identified by ultra-high performance liquid chromatography-quadrupole electrostatic field orbitrap high-resolution mass spectrometry(UPLC-Orbitrap-HRMS). The fragmentation pattern of the representative chemical compounds was summarized, and the potential anti-liver fibrosis active compounds of G. lucidum acting on the farnesoid X receptor(FXR) target were studied to elucidate its pharmacodynamic substance basis. Preliminarily, 95 chemical constituents of G. lucidum ethanol extracts were identified, including 24 ganoderic acids, 9 ganoderenic acids, 13 lucidenic acids, 3 ganolucidic acids, 1 ganoderma lactone, 40 other triterpenoids, 4 fatty acids, and 1 other constituent. In addition, the fragmentation patterns of the representative compounds were also analyzed. The structural characteristics of ganoderic acids and ganoderenic acids were the C30 skeleton, containing free-COOH and-OH groups, which could easily lose H_2O and CO_2 to form fragment ions. The D-ring was mostly a five-membered ring, which was prone to breakage. Lucidenic acids were the lanosterolane-type of the C27 skeleton, and the side-chain structure became shorter and contained the same free-COOH and-OH compared with ganoderic acids, which had been reduced from 8 to 5 cartons and prone to lose H_2O and CO_2. Then, six reported FXR receptor agonists were selected to form a training set for establishing a pharmacophore model based on FXR ligands. The 95 identified chemical constituents of G. lucidum were matched with the pharmacophore, and the optimal pharmacophore model 02(sensitivity=0.750 00, specificity=0.555 56, ROC=0.750) was selected for the virtual screening of the G. lucidum compound library through the validation of the test set. Finally, 31 potential G. lucidum active constituents were screened and chosen to activate the FXRs. The ADMET results showed that ganoderic acid H and lucidenic acid J had less than 90% plasma protein binding rate and no hepatotoxicity, which could be used as FXR activators for developing clinical drugs for the treatment of liver fibrosis, either alone or in combination.


Sujet(s)
Médicaments issus de plantes chinoises , Cirrhose du foie , Récepteurs cytoplasmiques et nucléaires , Reishi , Récepteurs cytoplasmiques et nucléaires/métabolisme , Récepteurs cytoplasmiques et nucléaires/composition chimique , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/métabolisme , Chromatographie en phase liquide à haute performance/méthodes , Humains , Reishi/composition chimique , Médicaments issus de plantes chinoises/composition chimique , Médicaments issus de plantes chinoises/pharmacologie , Spectrométrie de masse/méthodes , Structure moléculaire , Simulation de docking moléculaire
2.
Nat Commun ; 15(1): 6506, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39090079

RÉSUMÉ

The lack of an appropriate preclinical model of metabolic dysfunction-associated steatotic liver disease (MASLD) that recapitulates the whole disease spectrum impedes exploration of disease pathophysiology and the development of effective treatment strategies. Here, we develop a mouse model (Streptozotocin with high-fat diet, STZ + HFD) that gradually develops fatty liver, metabolic dysfunction-associated steatohepatitis (MASH), hepatic fibrosis, and hepatocellular carcinoma (HCC) in the context of metabolic dysfunction. The hepatic transcriptomic features of STZ + HFD mice closely reflect those of patients with obesity accompanying type 2 diabetes mellitus, MASH, and MASLD-related HCC. Dietary changes and tirzepatide administration alleviate MASH, hepatic fibrosis, and hepatic tumorigenesis in STZ + HFD mice. In conclusion, a murine model recapitulating the main histopathologic, transcriptomic, and metabolic alterations observed in MASLD patients is successfully established.


Sujet(s)
Carcinome hépatocellulaire , Alimentation riche en graisse , Modèles animaux de maladie humaine , Tumeurs du foie , Animaux , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/génétique , Mâle , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Souris , Alimentation riche en graisse/effets indésirables , Souris de lignée C57BL , Humains , Foie/métabolisme , Foie/anatomopathologie , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Streptozocine , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Transcriptome , Obésité/métabolisme , Obésité/complications , Diabète de type 2/complications , Diabète de type 2/métabolisme , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/complications
3.
J Biochem Mol Toxicol ; 38(8): e23788, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39087918

RÉSUMÉ

In this study, we evaluated the hepatoprotective effects of astaxanthin, a natural carotenoid, against the cholestatic liver fibrosis induced by bile duct ligation (BDL). Toward this end, male rats were subjected to BDL and treated with astaxanthin for 35 days. Afterwards, their serum and liver biochemical factors were assessed. Also, histopathological and immunohistochemical analyses were performed to determine the fibrosis and the expression levels of alpha-smooth muscle actin (α-SMA) and transforming growth factor beta (TGF-ß1) in the liver tissue. Based on the results, BDL caused a significant increase in liver enzyme levels, blood lipids, and bilirubin, while decreasing the activity of superoxide dismutase(SOD), catalase (CAT), and glutathione (GSH) enzymes. Also, in the BDL rats, hepatocyte necrosis, infiltration of inflammatory lymphocytes, and hyperplasia of bile ducts were detected, along with a significant increase in α-SMA and TGF-ß1 expression. Astaxanthin, however, significantly prevented the BDL's detrimental effects. In all, 10 mg/kg of this drug maintained the bilirubin and cholesterol serum levels of BDL rats at normal levels. It also reduced the liver enzymes' activity and serum lipids, while increasing the SOD, CAT, and GSH activity in BDL rats. The expression of α-SMA and TGF-ß1 in the BDL rats treated with 10 mg/kg of astaxanthin was moderate (in 34%-66% of cells) and no considerable cholestatic fibrosis was observed in this group. However, administrating the 20 mg/kg of astaxanthin was not effective in this regard. These findings showed that astaxanthin could considerably protect the liver from cholestatic damage by improving the biochemical features and regulating the expression of related proteins.


Sujet(s)
Conduits biliaires , Cholestase , Cirrhose du foie , Rat Wistar , Xanthophylles , Animaux , Xanthophylles/pharmacologie , Xanthophylles/usage thérapeutique , Mâle , Rats , Cholestase/anatomopathologie , Cholestase/métabolisme , Cholestase/traitement médicamenteux , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/prévention et contrôle , Ligature , Conduits biliaires/chirurgie , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Foie/métabolisme , Facteur de croissance transformant bêta-1/métabolisme
4.
PLoS One ; 19(8): e0308334, 2024.
Article de Anglais | MEDLINE | ID: mdl-39133714

RÉSUMÉ

Environmental pollutants, including polychlorinated biphenyls (PCBs), act as endocrine disruptors and impair various physiological processes. PCB 126 is associated with steatohepatitis, fibrosis, cirrhosis, and other hepatic injuries. These disorders can be regulated by microRNAs (miRNAs). Therefore, this study aimed to investigate the role of miRNAs in non-alcoholic fatty liver disease associated with exposure to PCB 126. Adult male C57BL/6 mice were exposed to PCB 126 (5 µmol/kg of body weight) for 10 weeks. The PCB group showed lipid accumulation in the liver in the presence of macro- and microvesicular steatosis and fibrosis with increased inflammatory and profibrotic gene expression, consistent with non-alcoholic steatohepatitis (NASH). PCB exposure also upregulated miR-155 and miR-34a, which induce the expression of proinflammatory cytokines and inflammation in the liver and reduce the expression of peroxisome proliferator-activated receptor α, which, in turn, impairs lipid oxidation and hepatic steatosis. Therefore, the present study showed that PCB 126 induced NASH via potential mechanisms involving miR-155 and miR-34a, which may contribute to the development of new diagnostic markers and therapeutic strategies.


Sujet(s)
Cirrhose du foie , Souris de lignée C57BL , microARN , Polychlorobiphényles , Régulation positive , Animaux , microARN/génétique , microARN/métabolisme , Polychlorobiphényles/toxicité , Mâle , Souris , Cirrhose du foie/induit chimiquement , Cirrhose du foie/génétique , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Régulation positive/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Stéatose hépatique non alcoolique/induit chimiquement , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/génétique , Stéatose hépatique non alcoolique/anatomopathologie , Polluants environnementaux/toxicité , Métabolisme lipidique/effets des médicaments et des substances chimiques , Métabolisme lipidique/génétique
5.
Int J Mol Sci ; 25(15)2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39126031

RÉSUMÉ

Nonalcoholic fatty liver disease (NAFLD), or metabolic dysfunction-associated steatotic liver disease (MASLD), is a liver condition that is linked to overweight, obesity, diabetes mellitus, and metabolic syndrome. Nonalcoholic steatohepatitis (NASH), or metabolic dysfunction-associated steatohepatitis (MASH), is a form of NAFLD/MASLD that progresses over time. While steatosis is a prominent histological characteristic and recognizable grossly and microscopically, liver biopsies of individuals with NASH/MASH may exhibit several other abnormalities, such as mononuclear inflammation in the portal and lobular regions, hepatocellular damage characterized by ballooning and programmed cell death (apoptosis), misfolded hepatocytic protein inclusions (Mallory-Denk bodies, MDBs), megamitochondria as hyaline inclusions, and fibrosis. Ballooning hepatocellular damage remains the defining feature of NASH/MASH. The fibrosis pattern is characterized by the initial expression of perisinusoidal fibrosis ("chicken wire") and fibrosis surrounding the central veins. Children may have an alternative form of progressive NAFLD/MASLD characterized by steatosis, inflammation, and fibrosis, mainly in Rappaport zone 1 of the liver acinus. To identify, synthesize, and analyze the scientific knowledge produced regarding the implications of using a score for evaluating NAFLD/MASLD in a comprehensive narrative review. The search for articles was conducted between 1 January 2000 and 31 December 2023, on the PubMed/MEDLINE, Scopus, Web of Science, and Cochrane databases. This search was complemented by a gray search, including internet browsers (e.g., Google) and textbooks. The following research question guided the study: "What are the basic data on using a score for evaluating NAFLD/MASLD?" All stages of the selection process were carried out by the single author. Of the 1783 articles found, 75 were included in the sample for analysis, which was implemented with an additional 25 articles from references and gray literature. The studies analyzed indicated the beneficial effects of scoring liver biopsies. Although similarity between alcoholic steatohepatitis (ASH) and NASH/MASH occurs, some patterns of hepatocellular damage seen in alcoholic disease of the liver do not happen in NASH/MASH, including cholestatic featuring steatohepatitis, alcoholic foamy degeneration, and sclerosing predominant hyaline necrosis. Generally, neutrophilic-rich cellular infiltrates, prominent hyaline inclusions and MDBs, cholestasis, and obvious pericellular sinusoidal fibrosis should favor the diagnosis of alcohol-induced hepatocellular injury over NASH/MASH. Multiple grading and staging methods are available for implementation in investigations and clinical trials, each possessing merits and drawbacks. The systems primarily used are the Brunt, the NASH CRN (NASH Clinical Research Network), and the SAF (steatosis, activity, and fibrosis) systems. Clinical investigations have utilized several approaches to link laboratory and demographic observations with histology findings with optimal platforms for clinical trials of rapidly commercialized drugs. It is promising that machine learning procedures (artificial intelligence) may be critical for developing new platforms to evaluate the benefits of current and future drug formulations.


Sujet(s)
Stéatose hépatique non alcoolique , Humains , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/métabolisme , Foie/anatomopathologie , Foie/métabolisme , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme
6.
Cell Mol Life Sci ; 81(1): 345, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39133305

RÉSUMÉ

BACKGROUND AND AIMS: Hepatitis B virus (HBV)-associated liver cirrhosis (LC), a common condition with high incidence and mortality rates, is often associated with diabetes mellitus (DM). However, the molecular mechanisms underlying impaired glucose regulation during HBV-associated LC remain unclear. METHODS: Data from 63 patients with LC and 62 patients with LC-associated DM were analysed. Co-culture of NK cells and islet ß cell lines were used to study the glucose regulation mechanism. A mouse model of LC was used to verify the effect of S100A8/A9 on the glucose regulation. RESULTS: Higher levels of interferon (IFN)-γ derived from natural killer (NK) cells and lower levels of insulin emerged in the peripheral blood of patients with both LC and DM compared with those from patients with LC only. IFN-γ derived from NK cells facilitated ß cell necroptosis and impaired insulin production. Furthermore, S100A8/A9 elevation in patients with both LC and DM was found to upregulate IFN-γ production in NK cells. Consistently, in the mouse model for LC, mice treated with carbon tetrachloride (CCL4) and S100A8/A9 exhibited increased blood glucose, impaired insulin production, increased IFN-γ, and increased ß cells necroptosis compared with those treated with CCL4. Mechanistically, S100A8/A9 activated the p38 MAPK pathway to increase IFN-γ production in NK cells. These effects were diminished after blocking RAGE. CONCLUSION: Together, the data indicate that IFN-γ produced by NK cells induces ß cell necroptosis via the S100A8/A9-RAGE-p38 MAPK axis in patients with LC and DM. Reduced levels of S100A8/A9, NK cells, and IFN-γ could be valuable for the treatment of LC with DM. Accumulation of S100A8/A9 in patients with LC may indicate the emergence of DM.


Sujet(s)
Calgranuline A , Calgranuline B , Virus de l'hépatite B , Cellules à insuline , Interféron gamma , Cellules tueuses naturelles , Cirrhose du foie , Nécroptose , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Humains , Animaux , Interféron gamma/métabolisme , Calgranuline B/métabolisme , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Cirrhose du foie/virologie , Cirrhose du foie/immunologie , Souris , Mâle , Cellules à insuline/métabolisme , Cellules à insuline/anatomopathologie , Cellules à insuline/virologie , Calgranuline A/métabolisme , Souris de lignée C57BL , Femelle , Adulte d'âge moyen , Hépatite B/complications , Hépatite B/anatomopathologie , Hépatite B/métabolisme , Modèles animaux de maladie humaine , Tétrachloro-méthane
7.
Commun Biol ; 7(1): 991, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39143151

RÉSUMÉ

Liver fibrosis progressing to cirrhosis is a major risk factor for liver cancer, impacting surgical treatment and survival. Our study focuses on the role of extracellular nicotinamide adenine dinucleotide (eNAD+) in liver fibrosis, analyzing liver disease patients undergoing surgery. Additionally, we explore NAD+'s therapeutic potential in a mouse model of extended liver resection and in vitro using 3D hepatocyte spheroids. eNAD+ correlated with aspartate transaminase (AST) and bilirubin after liver resection (AST: r = 0.2828, p = 0.0087; Bilirubin: r = 0.2584, p = 0.0176). Concordantly, post-hepatectomy liver failure (PHLF) was associated with higher eNAD+ peaks (n = 10; p = 0.0063). Post-operative eNAD+ levels decreased significantly (p < 0.05), but in advanced stages of liver fibrosis or cirrhosis, this decline not only diminished but actually showed a trend towards an increase. The expression of NAD+ biosynthesis rate-limiting enzymes, nicotinamide phosphoribosyltransferase (NAMPT) and nicotinamide mononucleotide adenylyltransferase 3 (NMNAT3), were upregulated significantly in the liver tissue of patients with higher liver fibrosis stages (p < 0.0001). Finally, the administration of NAD+ in a 3D hepatocyte spheroid model rescued hepatocytes from TNFalpha-induced cell death and improved viability (p < 0.0001). In a mouse model of extended liver resection, NAD+ treatment significantly improved survival (p = 0.0158) and liver regeneration (p = 0.0186). Our findings reveal that eNAD+ was upregulated in PHLF, and rate-limiting enzymes of NAD+ biosynthesis demonstrated higher expressions under liver fibrosis. Further, eNAD+ administration improved survival after extended liver resection in mice and enhanced hepatocyte viability in vitro. These insights may offer a potential target for future therapies.


Sujet(s)
Hépatectomie , Défaillance hépatique , NAD , NAD/métabolisme , Animaux , Humains , Souris , Défaillance hépatique/étiologie , Défaillance hépatique/métabolisme , Défaillance hépatique/anatomopathologie , Défaillance hépatique/chirurgie , Mâle , Hépatocytes/métabolisme , Adulte d'âge moyen , Femelle , Souris de lignée C57BL , Cirrhose du foie/métabolisme , Cirrhose du foie/chirurgie , Modèles animaux de maladie humaine , Sujet âgé
8.
Front Immunol ; 15: 1425384, 2024.
Article de Anglais | MEDLINE | ID: mdl-39139565

RÉSUMÉ

Background: Schistosomiasis is a zoonotic parasitic disorder induced by the infestation of schistosomes, a genus of trematodes. MicroRNAs (miRNAs) in egg-derived exosomes are crucial for modulating the host's immune responses and orchestrating the pathophysiological mechanisms. Although the exosomes secreted by S. japonicum contain abundant miRNAs, the specific roles of these miRNAs in the pathogenesis of schistosomiasis-induced hepatic fibrosis are yet to be comprehensively elucidated. The egg exosomes of S. japonicum secrete miRNA-30, a novel miRNA. Methods: In vitro, the effect of miRNA-30 was evaluated by transfecting HSCs with miRNA mimics. The target gene biosignature for miRNA-30 was predicted using the miRDB software. The effect of miRNA-30 in hepatic fibrosis was evaluated by either elevating its expression in healthy mice or by inhibiting its activity in infected mice by administration of recombinant adeno-associated virus serotype eight vectors expressing miRNA-30 or miRNA sponges. Results: This novel miRNA can activate hepatic stellate cells (HSCs), the prinary effector cells of hepatic fibrosis, in vitro, i.e., it significantly increases the fibrogenic factors Col1(α1), Col3(α1), and α-SMA at both mRNA and protein levels. In addition, miRNA-30 may activate HSCs by targeting the host RORA gene. In addition, in vivo experiments were conducted by administering a recombinant adeno-associated viral vector to modulate the expression levels of miRNA-30. The overexpression of miRNA-30 in healthy mice significantly elevated the expression of Col1(α1), Col3(α1), and α-SMA at both the transcriptomic and proteomic scales. This overexpression was coupled with a pronounced augmentation in the hepatic hydroxyproline content. Conversely, the in vivo silencing of miRNA-30 in infected mice induced a considerable reduction in the size of hepatic granulomas and areas of collagen deposition. Hence, in vivo, modulation of miRNA-30 expression may play a pivotal role in ameliorating the severity of hepatic fibrosis in mice afflicted with S. japonica. Conclusions: The study results suggest that miRNA-30 may augment schistosomiasis-induced hepatic fibrosis through a probable interaction with the host RORA. Our study may improve the current theoretical framework regarding cross-species regulation by miRNAs of hepatic fibrosis in schistosomiasis.


Sujet(s)
Cellules étoilées du foie , Cirrhose du foie , microARN , Schistosoma japonicum , Schistosomiase artérioveineuse , Animaux , microARN/génétique , Cirrhose du foie/parasitologie , Cirrhose du foie/génétique , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Souris , Schistosomiase artérioveineuse/immunologie , Schistosomiase artérioveineuse/génétique , Schistosomiase artérioveineuse/parasitologie , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/parasitologie , Exosomes/métabolisme , Exosomes/génétique , Femelle , Modèles animaux de maladie humaine , Ovule/métabolisme
9.
FASEB J ; 38(15): e23495, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39126242

RÉSUMÉ

Hepatic stellate cell (HSC) activation is the essential pathological process of liver fibrosis (LF). The molecular mechanisms regulating HSC activation and LF are incompletely understood. Here, we explored the effect of transcription factor SRY-related high mobility group box 7 (SOX7) on HSC activation and LF, and the underlying molecular mechanism. We found the expression levels of SOX7 were decreased in human and mouse fibrotic livers, particularly at the fibrotic foci. SOX7 was also downregulated in primary activated HSCs and TGF-ß1 stimulated LX-2 cells. SOX7 knockdown promoted activation and proliferation of LX-2 cells while inhibiting their apoptosis. On the other hand, overexpression of SOX7 suppressed the activation and proliferation of HSCs. Mechanistically, SOX7 attenuates HSC activation and LF by decreasing the expression of ß-catenin and phosphorylation of Smad2 and Smad3 induced by TGF-ß1. Furthermore, overexpression of SOX7 using AAV8-SOX7 mouse models ameliorated the extent of LF in response to CCl4 treatment in vivo. Collectively, SOX7 suppressed HSC activation and LF. Targeting SOX7, therefore, could be a potential novel strategy to protect against LF.


Sujet(s)
Cellules étoilées du foie , Cirrhose du foie , Facteurs de transcription SOX-F , Cellules étoilées du foie/métabolisme , Animaux , Cirrhose du foie/métabolisme , Cirrhose du foie/génétique , Cirrhose du foie/anatomopathologie , Souris , Humains , Mâle , Facteurs de transcription SOX-F/métabolisme , Facteurs de transcription SOX-F/génétique , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique , Prolifération cellulaire , Souris de lignée C57BL , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Apoptose , Protéine Smad2/métabolisme , Protéine Smad2/génétique , Lignée cellulaire , Protéine Smad-3/métabolisme , Protéine Smad-3/génétique
10.
Nutrients ; 16(15)2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39125267

RÉSUMÉ

The VLCKD is a diet recognized to promote rapid fat mobilization and reduce inflammation, hepatic steatosis, and liver fibrosis. Extracellular vesicles (EVs) mediate cell-to-cell communication. The aim of the study is to investigate the role of circulating EVs in cell proliferation, ketone bodies, and ROS production in patients on an 8-week VLCKD regimen. Participants were classified as responders (R) or non-responders (NR) to VLCKD treatment based on their fibroscan results. In vitro experiments with the hepatic cell lines HEPA-RG (normal hepatocytes) and LX-2 (stellate cells) were conducted to investigate the effects of circulating EVs on cell viability, ROS production, and ketone body presence. The findings reveal a notable reduction in cell viability in both cell lines when treated with exosomes (EXOs). In contrast, treatment with microvesicles (MVs) did not appear to affect cell viability, which remained unchanged. Additionally, the levels of ketone bodies measured in urine were not consistently correlated with the reduction of fibrosis in responders (R). Similarly, an increase in ketone bodies was observed in non-responders (NR), which was also not aligned with the expected reduction in fibrosis. This inconsistency stands in stark contrast to the levels of Reactive Oxygen Species (ROS), which exhibited a clear and consistent pattern in accordance with the dietary intervention. Finally, in this preliminary study, ROS has been identified as a potential diet adherence marker for VLCKD patients; the ROS levels reliably follow the progression of the fibrosis response, providing a more accurate reflection of the therapeutic effects.


Sujet(s)
Survie cellulaire , Régime cétogène , Vésicules extracellulaires , Hépatocytes , Corps cétoniques , Espèces réactives de l'oxygène , Humains , Espèces réactives de l'oxygène/métabolisme , Régime cétogène/méthodes , Vésicules extracellulaires/métabolisme , Mâle , Femelle , Corps cétoniques/métabolisme , Hépatocytes/métabolisme , Adulte , Adulte d'âge moyen , Lignée cellulaire , Cirrhose du foie/métabolisme , Cirrhose du foie/diétothérapie , Exosomes/métabolisme
11.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39125617

RÉSUMÉ

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


Sujet(s)
Hépatocytes , Facteur-2 apparenté à NF-E2 , Animaux , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Souris , Hépatocytes/métabolisme , Mâle , Femelle , Évolution de la maladie , Souris de lignée C57BL , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Stéatose hépatique/génétique , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Cirrhose du foie/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Facteur-1 apparenté à NF-E2/métabolisme , Facteur-1 apparenté à NF-E2/génétique , Facteur nucléaire-1 respiratoire/métabolisme , Facteur nucléaire-1 respiratoire/génétique , Alimentation riche en graisse/effets indésirables , Foie/métabolisme , Foie/anatomopathologie , Humains
12.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39125684

RÉSUMÉ

In addition to direct damage to hepatocytes, long-term ethanol consumption leads to lipid accumulation and hepatic steatosis, as well as to the dysregulation of lipid metabolism. The final step in various liver diseases is cirrhosis. The aim of this study was to compare the FA (fatty acids) profile and expression levels of genes involved in lipid metabolism in cirrhotic liver tissue and normal liver tissue. Exploring the changes in the FA profile and expression of genes related to fatty acid metabolism in cirrhotic liver tissue reveals a molecular landscape that goes beyond the surface of traditional liver function assessments. Understanding the shifts in gene expression and fatty acid composition in liver tissue opens avenues for interventions that may aid in the treatment of cirrhosis in the future.


Sujet(s)
Acides gras , Métabolisme lipidique , Cirrhose du foie , Foie , Acides gras/métabolisme , Cirrhose du foie/métabolisme , Cirrhose du foie/génétique , Humains , Métabolisme lipidique/génétique , Foie/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Régulation de l'expression des gènes , Analyse de profil d'expression de gènes , Sujet âgé
13.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39125751

RÉSUMÉ

Bilirubin plays a key role in early diagnosis, prognosis, and prevention of liver diseases. Unconjugated bilirubin (UCB) requires conversion to a water-soluble form through liver glucuronidation, producing monoglucuronide (BMG) or diglucuronide bilirubin (BDG) for bile excretion. This study aimed to assess the roles of bilirubin's molecular species-UCB, BMG, and BDG-in diagnosing and understanding the pathogenesis of liver cirrhosis in patients with acute-on-chronic liver failure (ACLF), compensated liver cirrhosis (LC) patients, and healthy individuals. The study included patients with ACLF and compensated LC of diverse etiologies, along with healthy controls. We collected laboratory and clinical data to determine the severity and assess mortality. We extracted bilirubin from serum samples to measure UCB, BMG, and BDG using liquid chromatography-mass spectrometry (LC-MS). The quantification of bilirubin was performed by monitoring the mass charge (m/z) ratio. Of the 74 patients assessed, 45 had ACLF, 11 had LC, and 18 were healthy individuals. Among ACLF patients, the levels of molecular species of bilirubin were UCB 19.69 µmol/L, BMG 47.71 µmol/L, and BDG 2.120 µmol/L. For compensated cirrhosis patients, the levels were UCB 11.29 µmol/L, BMG 1.49 µmol/L, and BDG 0.055 µmol/L, and in healthy individuals, the levels were UCB 6.42 µmol/L, BMG 0.52 µmol/L, and BDG 0.028 µmol/L. The study revealed marked elevations in the bilirubin species in individuals with ACLF compared to those with compensated cirrhosis and healthy controls, underscoring the progression of liver dysfunction. The correlation of BMG and BDG levels with commonly used inflammatory markers suggests a relationship between bilirubin metabolism and systemic inflammation in ACLF.


Sujet(s)
Insuffisance hépatique aigüe sur chronique , Bilirubine , Cirrhose du foie , Humains , Insuffisance hépatique aigüe sur chronique/métabolisme , Insuffisance hépatique aigüe sur chronique/sang , Insuffisance hépatique aigüe sur chronique/étiologie , Bilirubine/métabolisme , Bilirubine/sang , Femelle , Mâle , Adulte d'âge moyen , Adulte , Cirrhose du foie/métabolisme , Cirrhose du foie/sang , Cirrhose du foie/complications , Marqueurs biologiques/sang , Sujet âgé , Études cas-témoins , Pronostic , Chromatographie en phase liquide
14.
Life Sci ; 353: 122934, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39089644

RÉSUMÉ

The review focused mainly on the pathogenesis of hepatogenous diabetes (HD) in liver cirrhosis (LC). This review reveals parallels between the mechanisms of metabolic dysfunction observed in LC and type II diabetes (T2DM), suggesting a shared pathway leading to HD. It underscores the role of insulin in HD pathogenesis, highlighting key factors such as insulin signaling, glucose metabolism, insulin resistance (IR), and the influence of adipocytes. Furthermore, the impact of adipose tissue accumulation, fatty acid metabolism, and pro-inflammatory cytokines like Tumor necrosis factor-α (TNF-α) on IR are discussed in the context of HD. Altered signaling pathways, disruptions in the endocrine system, liver inflammation, changes in muscle mass and composition, and modifications to the gut microbiota collectively contribute to the complex interplay linking cirrhosis and HD. This study highlights how important it is to identify and treat this complex condition in cirrhotic patients by thoroughly analyzing the link between cirrhosis, IR, and HD. It also emphasizes the vitality of targeted interventions. Cellular and molecular investigations into IR have revealed potential therapeutic targets for managing and preventing HD.


Sujet(s)
Diabète de type 2 , Insulinorésistance , Cirrhose du foie , Humains , Cirrhose du foie/métabolisme , Insulinorésistance/physiologie , Diabète de type 2/métabolisme , Animaux , Transduction du signal , Insuline/métabolisme
15.
Front Immunol ; 15: 1428551, 2024.
Article de Anglais | MEDLINE | ID: mdl-39086479

RÉSUMÉ

Background: Myocardial inflammation and apoptosis induced by cirrhosis are among the primary mechanisms of cirrhotic cardiomyopathy. CD73, a common extracellular nucleotidase also known as 5'-nucleotidase, is associated with the progression of inflammation and immunity in multiple organs. However, the mechanism by which CD73 contributes to myocardial inflammation and apoptosis in cirrhosis remains unclear. Methods: In this study, a cirrhotic cardiomyopathy model in mice was established by bile duct ligation. Myocardial-specific overexpression of CD73 was achieved by tail vein injection of AAV9 (adeno-associated virus)-cTNT-NT5E-mCherry, and cardiac function in mice was assessed using echocardiography. Myocardial inflammation infiltration and apoptosis were evaluated through pathological observation and ELISA assays. The expression of CD73, A2AR, apoptotic markers, and proteins related to the NF-κB pathway in myocardial tissue were measured. Results: In the myocardial tissue of the cirrhotic cardiomyopathy mouse model, the expression of CD73 and A2AR increased. Overexpression of CD73 in the myocardium via AAV9 injection and stimulation of A2AR with CGS 21680 inhibited myocardial inflammation and cardiomyocyte apoptosis induced by cirrhosis. Additionally, overexpression of CD73 suppressed the activation of the NF-κB pathway by upregulating the expression of the adenosine receptor A2A. Conclusion: Our study reveals that the CD73/A2AR signaling axis mitigates myocardial inflammation and apoptosis induced by cirrhosis through negative feedback regulation of the NF-κB pathway.


Sujet(s)
5'-Nucleotidase , Cardiomyopathies , Cirrhose du foie , Récepteur A2A à l'adénosine , Transduction du signal , Animaux , Mâle , Souris , 5'-Nucleotidase/métabolisme , Apoptose , Cardiomyopathies/métabolisme , Cardiomyopathies/étiologie , Cardiomyopathies/immunologie , Modèles animaux de maladie humaine , Rétrocontrôle physiologique , Protéines liées au GPI , Cirrhose du foie/immunologie , Cirrhose du foie/métabolisme , Souris de lignée C57BL , Facteur de transcription NF-kappa B/métabolisme , Récepteur A2A à l'adénosine/métabolisme
16.
Sci Rep ; 14(1): 18840, 2024 08 13.
Article de Anglais | MEDLINE | ID: mdl-39138336

RÉSUMÉ

The combination of lineage tracing and immunohistochemistry has helped to identify subpopulations and fate of hepatic stellate cells (HSC) in murine liver. HSC are sinusoidal pericytes that act as myofibroblast precursors after liver injury. Single cell RNA sequencing approaches have recently helped to differentiate central and portal HSC. A specific Cre line to lineage trace portal HSC has not yet been described. We used three Cre lines (Lrat-Cre, PDGFRß-CreERT2 and SMMHC-CreERT2) known to label mesenchymal cells including HSC in combination with a tdTomato-expressing reporter. All three Cre lines labeled populations of HSC as well as smooth muscle cells (SMC). Using the SMMHC-CreERT2, we identified a subtype of HSC in the periportal area of the hepatic lobule (termed zone 1-HSC). We lineage traced tdTomato-expressing zone 1-HSC over 1 year, described fibrotic behavior in two fibrosis models and investigated their possible role during fibrosis. This HSC subtype resides in zone 1 under healthy conditions; however, zonation is disrupted in preclinical models of liver fibrosis (CCl4 and MASH). Zone 1-HSC do not transform into αSMA-expressing myofibroblasts. Rather, they participate in sinusoidal capillarization. We describe a novel subtype of HSC restricted to zone 1 under physiological conditions and its possible function after liver injury. In contrast to the accepted notion, this HSC subtype does not transform into αSMA-positive myofibroblasts; rather, zone 1-HSC adopt properties of capillary pericytes, thereby participating in sinusoidal capillarization.


Sujet(s)
Cellules étoilées du foie , Cirrhose du foie , Myofibroblastes , Animaux , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/anatomopathologie , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Souris , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Foie/anatomopathologie , Foie/métabolisme , Péricytes/métabolisme , Péricytes/anatomopathologie , Lignage cellulaire , Mâle , Différenciation cellulaire , Modèles animaux de maladie humaine , Souris de lignée C57BL
17.
World J Gastroenterol ; 30(28): 3428-3446, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-39091710

RÉSUMÉ

BACKGROUND: Alcohol-associated liver disease (ALD) is a leading cause of liver-related morbidity and mortality, but there are no therapeutic targets and modalities to prevent ALD-related liver fibrosis. Peroxisome proliferator activated receptor (PPAR) α and δ play a key role in lipid metabolism and intestinal barrier homeostasis, which are major contributors to the pathological progression of ALD. Meanwhile, elafibranor (EFN), which is a dual PPARα and PPARδ agonist, has reached a phase III clinical trial for the treatment of metabolic dysfunction-associated steatotic liver disease and primary biliary cholangitis. However, the benefits of EFN for ALD treatment is unknown. AIM: To evaluate the inhibitory effects of EFN on liver fibrosis and gut-intestinal barrier dysfunction in an ALD mouse model. METHODS: ALD-related liver fibrosis was induced in female C57BL/6J mice by feeding a 2.5% ethanol (EtOH)-containing Lieber-DeCarli liquid diet and intraperitoneally injecting carbon tetrachloride thrice weekly (1 mL/kg) for 8 weeks. EFN (3 and 10 mg/kg/day) was orally administered during the experimental period. Histological and molecular analyses were performed to assess the effect of EFN on steatohepatitis, fibrosis, and intestinal barrier integrity. The EFN effects on HepG2 lipotoxicity and Caco-2 barrier function were evaluated by cell-based assays. RESULTS: The hepatic steatosis, apoptosis, and fibrosis in the ALD mice model were significantly attenuated by EFN treatment. EFN promoted lipolysis and ß-oxidation and enhanced autophagic and antioxidant capacities in EtOH-stimulated HepG2 cells, primarily through PPARα activation. Moreover, EFN inhibited the Kupffer cell-mediated inflammatory response, with blunted hepatic exposure to lipopolysaccharide (LPS) and toll like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) signaling. EFN improved intestinal hyperpermeability by restoring tight junction proteins and autophagy and by inhibiting apoptosis and proinflammatory responses. The protective effect on intestinal barrier function in the EtOH-stimulated Caco-2 cells was predominantly mediated by PPARδ activation. CONCLUSION: EFN reduced ALD-related fibrosis by inhibiting lipid accumulation and apoptosis, enhancing hepatocyte autophagic and antioxidant capacities, and suppressing LPS/TLR4/NF-κB-mediated inflammatory responses by restoring intestinal barrier function.


Sujet(s)
Chalcones , Modèles animaux de maladie humaine , Muqueuse intestinale , Cirrhose du foie , Maladies alcooliques du foie , Souris de lignée C57BL , Récepteur PPAR alpha , Animaux , Souris , Humains , Femelle , Maladies alcooliques du foie/anatomopathologie , Maladies alcooliques du foie/métabolisme , Maladies alcooliques du foie/prévention et contrôle , Maladies alcooliques du foie/étiologie , Maladies alcooliques du foie/traitement médicamenteux , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/agonistes , Chalcones/pharmacologie , Cirrhose du foie/anatomopathologie , Cirrhose du foie/induit chimiquement , Cirrhose du foie/métabolisme , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/prévention et contrôle , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/métabolisme , Cellules Caco-2 , Foie/anatomopathologie , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Éthanol/toxicité , Apoptose/effets des médicaments et des substances chimiques , Métabolisme lipidique/effets des médicaments et des substances chimiques , Récepteur PPAR delta/agonistes , Récepteur PPAR delta/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Propionates
18.
Clin Transl Med ; 14(8): e1793, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39113232

RÉSUMÉ

INTRODUCTION: Liver fibrosis is primarily driven by the activation of hepatic stellate cells (HSCs), which involves various epigenetic modifications. OBJECTIVES: N6-methyladenosine (m6A), the most prevalent RNA modification in eukaryotic cells, influences numerous physiological and pathological processes. Nevertheless, the role of insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3), a reader gene mediating m6A modifications, in liver fibrosis remains unclear. METHODS AND RESULTS: This study demonstrated that IGF2BP3 knockout reduces liver fibrosis by promoting HSC ferroptosis (FPT) and inactivating HSCs. Multi-omics analysis revealed that HSC-specific IGF2BP3 knockout decreased m6A content in Jagged1 (Jag1), a key component of the Notch signalling pathway. Furthermore, IGF2BP3 deficiency significantly reduced the expression of hairy and enhancer of split-1 (Hes1), a transcription factor in the Notch/Jag1 signalling pathway, with mRNA levels declining to 35%-62% and protein levels to 28%-35%. Additionally, it suppressed glutathione peroxidase 4 (GPX4) (decreased to approximately 31%-38%), a negative regulator of FPT, thereby facilitating HSC FPT progression and reducing profibrotic gene expression. CONCLUSION: These findings uncover a novel IGF2BP3/Notch/Jag1 signalling pathway involving HSC FPT, suggesting promising targets for ameliorating liver fibrosis. KEY POINTS/HIGHLIGHTS: IGF2BP3 deficiency inactivates Jag1 signalling. IGF2BP3 deficiency-mediated m6A modifications promote HSC ferroptosis. IGF2BP3 inhibition facilitates ferroptosis in HSCs via the Hes1/GPX4 axis. IGF2BP3 deficiency inactivates Jag1/Notch1/3/Hes1 signalling pathway inactivation, leading to the decrease in GPX4, which contributes to HSC ferroptosis.


Sujet(s)
Ferroptose , Cellules étoilées du foie , Protéine jagged-1 , Cirrhose du foie , Protéines de liaison à l'ARN , Récepteurs Notch , Transduction du signal , Ferroptose/génétique , Cellules étoilées du foie/métabolisme , Animaux , Cirrhose du foie/génétique , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Souris , Protéine jagged-1/génétique , Protéine jagged-1/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Transduction du signal/génétique , Récepteurs Notch/métabolisme , Récepteurs Notch/génétique , Souris knockout , Mâle , Humains
19.
Mol Med ; 30(1): 104, 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39030473

RÉSUMÉ

Cholestatic liver diseases (CLD) are characterized by impaired normal bile flow, culminating in excessive accumulation of toxic bile acids. The majority of patients with CLD ultimately progress to liver cirrhosis and hepatic failure, necessitating liver transplantation due to the lack of effective treatment. Recent investigations have underscored the pivotal role of the gut microbiota-bile acid axis in the progression of hepatic fibrosis via various pathways. The obstruction of bile drainage can induce gut microbiota dysbiosis and disrupt the intestinal mucosal barrier, leading to bacteria translocation. The microbial translocation activates the immune response and promotes liver fibrosis progression. The identification of therapeutic targets for modulating the gut microbiota-bile acid axis represents a promising strategy to ameliorate or perhaps reverse liver fibrosis in CLD. This review focuses on the mechanisms in the gut microbiota-bile acids axis in CLD and highlights potential therapeutic targets, aiming to lay a foundation for innovative treatment approaches.


Sujet(s)
Acides et sels biliaires , Cholestase , Dysbiose , Microbiome gastro-intestinal , Humains , Acides et sels biliaires/métabolisme , Animaux , Cholestase/métabolisme , Cholestase/microbiologie , Maladies du foie/métabolisme , Maladies du foie/microbiologie , Maladies du foie/étiologie , Cirrhose du foie/métabolisme , Cirrhose du foie/microbiologie
20.
Nat Commun ; 15(1): 5506, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951527

RÉSUMÉ

Obesity is a major cause of metabolic dysfunction-associated steatohepatitis (MASH) and is characterized by inflammation and insulin resistance. Interferon-γ (IFNγ) is a pro-inflammatory cytokine elevated in obesity and modulating macrophage functions. Here, we show that male mice with loss of IFNγ signaling in myeloid cells (Lyz-IFNγR2-/-) are protected from diet-induced insulin resistance despite fatty liver. Obesity-mediated liver inflammation is also attenuated with reduced interleukin (IL)-12, a cytokine primarily released by macrophages, and IL-12 treatment in vivo causes insulin resistance by impairing hepatic insulin signaling. Following MASH diets, Lyz-IFNγR2-/- mice are rescued from developing liver fibrosis, which is associated with reduced fibroblast growth factor (FGF) 21 levels. These results indicate critical roles for IFNγ signaling in macrophages and their release of IL-12 in modulating obesity-mediated insulin resistance and fatty liver progression to MASH. In this work, we identify the IFNγ-IL12 axis in regulating intercellular crosstalk in the liver and as potential therapeutic targets to treat MASH.


Sujet(s)
Stéatose hépatique , Insulinorésistance , Interféron gamma , Interleukine-12 , Foie , Macrophages , Souris knockout , Obésité , Transduction du signal , Animaux , Interféron gamma/métabolisme , Interleukine-12/métabolisme , Mâle , Obésité/métabolisme , Souris , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Macrophages/métabolisme , Foie/métabolisme , Foie/anatomopathologie , Souris de lignée C57BL , Alimentation riche en graisse/effets indésirables , Récepteur interféron/métabolisme , Récepteur interféron/génétique , , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Cirrhose du foie/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE