Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.556
Filtrer
1.
Sci Rep ; 14(1): 15246, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956068

RÉSUMÉ

This study aimed to explore the effects of peroxisome proliferator-activated receptor α (PPAR-α), a known inhibitor of ferroptosis, in Myocardial ischemia/reperfusion injury (MIRI) and its related mechanisms. In vivo and in vitro MIRI models were established. Our results showed that activation of PPAR-α decreased the size of the myocardial infarct, maintained cardiac function, and decreased the serum contents of creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH), and Fe2+ in ischemia/reperfusion (I/R)-treated mice. Additionally, the results of H&E staining, DHE staining, TUNEL staining, and transmission electron microscopy demonstrated that activation of PPAR-α inhibited MIRI-induced heart tissue and mitochondrial damage. It was also found that activation of PPAR-α attenuated MIRI-induced ferroptosis as shown by a reduction in malondialdehyde, total iron, and reactive oxygen species (ROS). In vitro experiments showed that intracellular contents of malondialdehyde, total iron, LDH, reactive oxygen species (ROS), lipid ROS, oxidized glutathione disulphide (GSSG), and Fe2+ were reduced by the activation of PPAR-α in H9c2 cells treated with anoxia/reoxygenation (A/R), while the cell viability and GSH were increased after PPAR-α activation. Additionally, changes in protein levels of the ferroptosis marker further confirmed the beneficial effects of PPAR-α activation on MIRI-induced ferroptosis. Moreover, the results of immunofluorescence and dual-luciferase reporter assay revealed that PPAR-α achieved its activity via binding to the 14-3-3η promoter, promoting its expression level. Moreover, the cardioprotective effects of PPAR-α could be canceled by pAd/14-3-3η-shRNA or Compound C11 (14-3-3η inhibitor). In conclusion, our results indicated that ferroptosis plays a key role in aggravating MIRI, and PPAR-α/14-3-3η pathway-mediated ferroptosis and mitochondrial injury might be an effective therapeutic target against MIRI.


Sujet(s)
Protéines 14-3-3 , Ferroptose , Lésion de reperfusion myocardique , Récepteur PPAR alpha , Ferroptose/effets des médicaments et des substances chimiques , Animaux , Récepteur PPAR alpha/métabolisme , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Protéines 14-3-3/métabolisme , Souris , Mâle , Espèces réactives de l'oxygène/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Lignée cellulaire , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Souris de lignée C57BL , Rats , Modèles animaux de maladie humaine
2.
FASEB J ; 38(13): e23788, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38963329

RÉSUMÉ

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


Sujet(s)
Cuivre , Ferroptose , Hypoxie , Souris de lignée C57BL , Animaux , Cuivre/métabolisme , Cuivre/déficit , Mâle , Souris , Hypoxie/métabolisme , Humains , Cellules HepG2 , Foie/métabolisme , Foie/anatomopathologie , Stress oxydatif , Métabolisme lipidique , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Stéatose hépatique/étiologie , Fer/métabolisme , Coenzyme A ligases/métabolisme , Coenzyme A ligases/génétique , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique
3.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39000217

RÉSUMÉ

Peroxisome proliferator-activated receptors (PPARs) may play an important role in the pathomechanism/pathogenesis of Alzheimer's disease (AD) and several other neurological/neuropsychiatric disorders. AD leads to progressive alterations in the redox state, ion homeostasis, lipids, and protein metabolism. Significant alterations in molecular processes and the functioning of several signaling pathways result in the degeneration and death of synapses and neuronal cells, leading to the most severe dementia. Peroxisome proliferator-activated receptor alpha (PPAR-α) is among the processes affected by AD; it regulates the transcription of genes related to the metabolism of cholesterol, fatty acids, other lipids and neurotransmission, mitochondria biogenesis, and function. PPAR-α is involved in the cholesterol transport to mitochondria, the substrate for neurosteroid biosynthesis. PPAR-α-coding enzymes, such as sulfotransferases, which are responsible for neurosteroid sulfation. The relation between PPAR-α and cholesterol/neurosteroids may have a significant impact on the course and progression of neurodegeneration/neuroprotection processes. Unfortunately, despite many years of intensive studies, the pathogenesis of AD is unknown and therapy for AD and other neurodegenerative diseases is symptomatic, presenting a significant goal and challenge today. This review presents recent achievements in therapeutic approaches for AD, which are targeting PPAR-α and its relation to cholesterol and neurosteroids in AD and neuropsychiatric disorders.


Sujet(s)
Maladie d'Alzheimer , Neurostéroïdes , Récepteur PPAR alpha , Animaux , Humains , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/traitement médicamenteux , Cholestérol/métabolisme , Troubles mentaux/métabolisme , Troubles mentaux/traitement médicamenteux , Mitochondries/métabolisme , Thérapie moléculaire ciblée , Neurostéroïdes/métabolisme , Récepteur PPAR alpha/métabolisme
4.
Toxicol Lett ; 398: 150-160, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38971454

RÉSUMÉ

Activation of pregnane X receptor (PXR) by xenobiotics has been associated with metabolic diseases. This study aimed to reveal the impact of PXR activation on hepatic metabolome and explore novel mechanisms underlying PXR-mediated lipid metabolism disorder in the liver. Wild-type and PXR-deficient male C57BL/6 mice were used as in vivo models, and hepatic steatosis was induced by pregnenolone-16α-carbonitrile, a typical rodent PXR agonist. Metabolomic analysis of liver tissues showed that PXR activation led to significant changes in metabolites involved in multiple metabolic pathways previously reported, including lipid metabolism, energy homeostasis, and amino acid metabolism. Moreover, the level of hepatic all-trans retinoic acid (ATRA), the main active metabolite of vitamin A, was significantly increased by PXR activation, and genes involved in ATRA metabolism exhibited differential expression following PXR activation or deficiency. Consistent with previous research, the expression of downstream target genes of peroxisome proliferator-activated receptor α (PPARα) was decreased. Analysis of fatty acids by Gas Chromatography-Mass Spectrometer further revealed changes in polyunsaturated fatty acid metabolism upon PXR activation, suggesting inhibition of PPARα activity. Taken together, our findings reveal a novel metabolomic signature of hepatic steatosis induced by PXR activation in mice.


Sujet(s)
Acides gras insaturés , Stéatose hépatique , Foie , Métabolomique , Souris de lignée C57BL , Récepteur PPAR alpha , Récepteur du prégnane X , Trétinoïne , Animaux , Mâle , Récepteur du prégnane X/métabolisme , Récepteur du prégnane X/génétique , Trétinoïne/métabolisme , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Stéatose hépatique/métabolisme , Stéatose hépatique/induit chimiquement , Acides gras insaturés/métabolisme , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique , Métabolisme lipidique/effets des médicaments et des substances chimiques , Souris , Souris knockout , Prégnénolone carbonitrile/pharmacologie , Modèles animaux de maladie humaine
5.
Front Biosci (Landmark Ed) ; 29(6): 209, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38940024

RÉSUMÉ

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) is a prevalent condition characterized by hepatic fat accumulation, often progressing to severe liver injury, for which approved treatments are currently lacking. This study explores the potential therapeutic impact of alpha-lipoic acid (ALA), a natural compound crucial in lipid metabolism, on NAFLD using an in vitro model. METHODS: HepG2 cells were treated with a palmitic acid:oleic acid (PA:OA) mixture, representing a cellular model of steatosis. Subsequent treatment with ALA at concentrations of 1 µM and 5 µM aimed to evaluate its effects on lipid content and metabolism. Real-time polymerase chain reaction (PCR), BODIPY staining, cytofluorimetric analysis, and lipidomics were used to assess gene expression, lipid droplet accumulation, and fatty acid profiles. RESULTS: Our results showed that ALA significantly reduced lipid droplets in PA:OA-treated HepG2 cells, with a concentration-dependent effect. Analysis of fatty acid profiles demonstrated a decrease in palmitic acid levels with ALA treatment, while oleic acid reduction was observed only at the higher concentration. Moreover, ALA modulated the expression of genes involved in cholesterol biosynthesis and low-density lipoprotein (LDL) metabolism, indicating a potential role in lipid homeostasis. Further insights into molecular mechanisms revealed that ALA modulated peroxisome proliferator activated receptors (PPARs), specifically PPAR-alpha and PPAR-gamma, involved in fatty acid metabolism and insulin sensitivity. Finally, ALA counteracted the overexpression of thermogenic genes induced by exogenous fatty acids, suggesting a regulatory role in energy dissipation pathways. CONCLUSION: In conclusion, this study highlights ALA as a therapeutic agent in mitigating lipid accumulation and dysregulation in NAFLD.


Sujet(s)
Métabolisme lipidique , Stéatose hépatique non alcoolique , Acide oléique , Acide palmitique , Acide lipoïque , Humains , Acide lipoïque/pharmacologie , Cellules HepG2 , Métabolisme lipidique/effets des médicaments et des substances chimiques , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/génétique , Acide oléique/pharmacologie , Acide oléique/métabolisme , Acide palmitique/pharmacologie , Acide palmitique/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Acides gras/métabolisme , Récepteur PPAR gamma/métabolisme , Gouttelettes lipidiques/métabolisme , Gouttelettes lipidiques/effets des médicaments et des substances chimiques , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique , Protéine-2 de découplage/métabolisme , Protéine-2 de découplage/génétique
6.
Eur J Med Chem ; 275: 116567, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-38865743

RÉSUMÉ

New analogs of the PPAR pan agonist AL29-26 encompassed ligand (S)-7 showing potent activation of PPARα and -γ subtypes as a partial agonist. In vitro experiments and docking studies in the presence of PPAR antagonists were performed to help interpretation of biological data and investigate the main interactions at the binding sites. Further in vitro experiments showed that (S)-7 induced anti-steatotic effects and enhancement of the glucose uptake. This latter effect could be partially ascribed to a significant inhibition of the mitochondrial pyruvate carrier demonstrating that (S)-7 also acted through insulin-independent mechanisms. In vivo experiments showed that this compound reduced blood glucose and lipid levels in a diabetic mice model displaying no toxicity on bone, kidney, and liver. To our knowledge, this is the first example of dual PPARα/γ partial agonist showing these combined effects representing, therefore, the potential lead of new drugs for treatment of dyslipidemic type 2 diabetes.


Sujet(s)
Hypoglycémiants , Récepteur PPAR alpha , Récepteur PPAR gamma , Animaux , Récepteur PPAR alpha/agonistes , Récepteur PPAR alpha/métabolisme , Récepteur PPAR gamma/agonistes , Récepteur PPAR gamma/métabolisme , Souris , Hypoglycémiants/pharmacologie , Hypoglycémiants/composition chimique , Hypoglycémiants/synthèse chimique , Humains , Relation structure-activité , Diabète expérimental/traitement médicamenteux , Diabète expérimental/métabolisme , Mâle , Structure moléculaire , Relation dose-effet des médicaments , Transporteurs d'acides monocarboxyliques/antagonistes et inhibiteurs , Transporteurs d'acides monocarboxyliques/métabolisme , Diabète de type 2/traitement médicamenteux , Diabète de type 2/métabolisme , Simulation de docking moléculaire , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme
7.
Discov Med ; 36(185): 1139-1153, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38926100

RÉSUMÉ

BACKGROUND: Metabolic dysfunction-associated steatotic liver disease (MASLD), and more specifically steatohepatitis may be associated with fat infiltration of skeletal muscles which is known as myosteatosis. Pan-peroxisome proliferator-activated receptor (PPAR) agonists have been shown to promote metabolic dysfunction-associated steatohepatitis (MASH) remission. However, the effect of PPAR agonists on myosteatosis remains to be determined. The aim of this review is to evaluate the effect that PPAR agonists alone or in combination, have on myosteatosis in the context of MASLD. METHODS: Original research reports concerning the impact of PPAR agonists on muscle fat in MASLD were screened from PUBMED and EMBASE databases following the PRISMA methodology. RESULTS: Eleven original manuscripts were included in this review. Two preclinical studies assessed the impact of the PPARα agonist on fat content in the quadriceps muscle and the liver by extracting triglycerides in rats fed a high-fat diet and in insulin-resistant mice. Both models showed muscle and liver triglyceride content reduction using WY14643. Fenofibrate had no significant impact on soleus intramyocellular lipids or liver fat content in insulin-resistant subjects based on proton magnetic resonance spectroscopy. Treatment with PPARδ agonists increased the expression of genes involved in fatty acid oxidation in two studies on muscle cell culture. PPARγ agonists were investigated in two preclinical studies and one clinical study using spectroscopy and computed tomography respectively. In the first preclinical study in Zucker diabetic fatty rats, rosiglitazone reduced muscle lipids and hepatic steatosis. In a second preclinical study using the same animal model, pioglitazone reduced tibialis anterior intramyocellular lipids. In contrast, computed tomography analyses in patients with type 2 diabetes revealed a surface area increase of low-density muscles (suggesting an increase in muscle fat content) after a one-year treatment with rosiglitazone. Varying combinations of PPAR agonists (cevoglitazar, fenofibrate/pioglitazone and muraglitazar) were evaluated in two preclinical studies and one clinical study. In rats, these treatments showed variable results for muscle and liver depending on the combinations studied. In type 2 diabetic patients, treatment with muraglitazar (a PPARα/γ agonist) reduced the intramyocellular lipid content of tibialis anterior as well as liver fat content following spectroscopy assessment. CONCLUSION: The combination of different PPAR agonists could have a positive impact on reducing myosteatosis, in addition to their effect on the liver. Some discrepancies could be explained by the different techniques used to assess muscle lipid content, the muscles assessed and the possible adipogenic effect of PPARγ agonists. Further clinical research is needed to fully assess the efficacy of these treatments on both MASLD progression and associated myosteatosis.


Sujet(s)
Stéatose hépatique , Animaux , Humains , Stéatose hépatique/traitement médicamenteux , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Récepteurs activés par les proliférateurs de peroxysomes/agonistes , Récepteurs activés par les proliférateurs de peroxysomes/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Rats , Souris , Récepteur PPAR alpha/agonistes , Récepteur PPAR alpha/métabolisme
8.
J Ethnopharmacol ; 333: 118487, 2024 Oct 28.
Article de Anglais | MEDLINE | ID: mdl-38925322

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease worldwide. Nonalcoholic steatohepatitis (NASH) is a crucial component of this disease spectrum. The Yanxiao Di'naer formula (YXDNE) is an Uyghur medical extract that has been used in folk medicine to treat hepatitis for a long time. However, the role and mechanism of action of YXDNE in NASH treatment remains unclear. OBJECTIVE: The objective of this study was to assess the effectiveness of YXDNE in treating NASH induced by injections of carbon tetrachloride combined with a high-fat high-cholesterol diet (HFHCD), and to clarify the underlying mechanisms. METHODS: The compounds in the YXDNE extract were analysed for classification and proportions using ultra-performance liquid chromatography-mass spectrometry. The efficacy of YXDNE in treating abnormal lipid metabolism was evaluated in L02 cells in vitro. In addition, a C57BL/6 mouse model of NASH was established to evaluate the therapeutic efficacy of YXDNE in vivo. Metabolomics and RNA sequencing were used to analyse the therapeutic effects of YXDNE on the liver. The corresponding signalling pathways were found to target AMPKα1, PPARα, and NF-κB. The efficacy of YXDNE was validated using inhibitors or silencing RNA (siRNA) against AMPKα1 and PPARα. RESULTS: This study confirmed that YXDNE treatment ameliorated NASH in a murine model of this disease. Metabolomics analysis suggested that YXDNE efficacy was associated with fatty acid catabolism and AMPK signalling pathways. RNA sequencing results showed that YXDNE efficacy in treating NASH was highly correlated with the AMPK, PPARα and NF-κB pathways. Both in vitro and in vivo experimental data demonstrated that YXDNE affected the expression of p-AMPKα1, PPARα, p-NF-κB, IκB, and p-IκB. The efficacy of YXDNE in treating NASH in vitro was cancelled when AMPK was inhibited with Compound C or PPARα was modulated via siRNA. CONCLUSIONS: YXDNE may have a therapeutic effect on abnormal lipid metabolism in L02 cells and in a murine model of NASH by affecting the AMPKα1/PPARα/NF-κB signalling pathway. Therefore, YXDNE has the potential for clinical application in the prevention and treatment of NASH.


Sujet(s)
Médicaments issus de plantes chinoises , Métabolomique , Souris de lignée C57BL , Stéatose hépatique non alcoolique , Animaux , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/métabolisme , Mâle , Souris , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/composition chimique , Médicaments issus de plantes chinoises/usage thérapeutique , Humains , Métabolisme lipidique/effets des médicaments et des substances chimiques , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique , Alimentation riche en graisse/effets indésirables , Modèles animaux de maladie humaine , Analyse de séquence d'ARN , Lignée cellulaire , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , AMP-Activated Protein Kinases/métabolisme
9.
Physiol Rep ; 12(12): e16117, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38898524

RÉSUMÉ

This study aimed to investigate how intermittent hyperoxic exposure (three cycles of 21% O2 [10 min] and 30% O2 [15 min]) affects exercise performance in mice. Three hours after the acute exposure, there was an observed increase in mRNA levels of phosphofructokinase (Bayes factor [BF] ≥ 10), mitochondrial transcription factor-A (BF ≥10), PPAR-α (BF ≥3), and PPAR-γ (BF ≥3) in the red gastrocnemius muscle (Gr). Four weeks of exercise training under intermittent (INT), but not continuous (HYP), hyperoxia significantly (BF ≥30) increased maximal exercise capacity compared to normoxic exercise-trained (ET) group. INT group exhibited significantly higher activity levels of 3-hydroxyacyl-CoA-dehydrogenase (HAD) in Gr (BF = 7.9) compared to ET group. Pyruvate dehydrogenase complex activity levels were significantly higher in INT group compared to ET group in white gastrocnemius, diaphragm, and left ventricle (BF ≥3). NT-PGC1α protein levels in Gr (BF = 7.7) and HAD activity levels in Gr (BF = 6.9) and soleus muscles (BF = 3.3) showed a significant positive correlation with maximal work values. These findings suggest that exercise training under intermittent hyperoxia is a beneficial strategy for enhancing endurance performance by improving fatty acid and pyruvic acid utilization.


Sujet(s)
Muscles squelettiques , Conditionnement physique d'animal , Endurance physique , Animaux , Mâle , Muscles squelettiques/métabolisme , Souris , Conditionnement physique d'animal/méthodes , Conditionnement physique d'animal/physiologie , Endurance physique/physiologie , Souris de lignée C57BL , Hyperoxie/métabolisme , Hyperoxie/physiopathologie , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Phosphofructokinases/métabolisme , Phosphofructokinases/génétique , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines de liaison à l'ADN , Protéines mitochondriales
10.
Sci Rep ; 14(1): 13435, 2024 06 11.
Article de Anglais | MEDLINE | ID: mdl-38862650

RÉSUMÉ

Diabetic corneal neuropathy (DCN) is a common diabetic ocular complication with limited treatment options. In this study, we investigated the effects of topical and oral fenofibrate, a peroxisome proliferator-activated receptor-α agonist, on the amelioration of DCN using diabetic mice (n = 120). Ocular surface assessments, corneal nerve and cell imaging analysis, tear proteomics and its associated biological pathways, immuno-histochemistry and western blot on PPARα expression, were studied before and 12 weeks after treatment. At 12 weeks, PPARα expression markedly restored after topical and oral fenofibrate. Topical fenofibrate significantly improved corneal nerve fibre density (CNFD) and tortuosity coefficient. Likewise, oral fenofibrate significantly improved CNFD. Both topical and oral forms significantly improved corneal sensitivity. Additionally, topical and oral fenofibrate significantly alleviated diabetic keratopathy, with fenofibrate eye drops demonstrating earlier therapeutic effects. Both topical and oral fenofibrate significantly increased corneal ß-III tubulin expression. Topical fenofibrate reduced neuroinflammation by significantly increasing the levels of nerve growth factor and substance P. It also significantly increased ß-III-tubulin and reduced CDC42 mRNA expression in trigeminal ganglions. Proteomic analysis showed that neurotrophin signalling and anti-inflammation reactions were significantly up-regulated after fenofibrate treatment, whether applied topically or orally. This study concluded that both topical and oral fenofibrate ameliorate DCN, while topical fenofibrate significantly reduces neuroinflammation.


Sujet(s)
Cornée , Diabète expérimental , Neuropathies diabétiques , Fénofibrate , Récepteur PPAR alpha , Animaux , Récepteur PPAR alpha/agonistes , Récepteur PPAR alpha/métabolisme , Souris , Fénofibrate/pharmacologie , Fénofibrate/administration et posologie , Diabète expérimental/traitement médicamenteux , Diabète expérimental/complications , Diabète expérimental/métabolisme , Neuropathies diabétiques/traitement médicamenteux , Neuropathies diabétiques/métabolisme , Cornée/métabolisme , Cornée/effets des médicaments et des substances chimiques , Cornée/innervation , Cornée/anatomopathologie , Mâle , Administration par voie orale , Administration par voie topique , Maladies de la cornée/traitement médicamenteux , Maladies de la cornée/étiologie , Maladies de la cornée/métabolisme , Maladies de la cornée/anatomopathologie , Souris de lignée C57BL , Protéomique/méthodes
11.
Life Sci ; 350: 122763, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38823505

RÉSUMÉ

AIMS: The intricate molecular mechanisms underlying estrogen receptor-positive (ER+) breast carcinogenesis and resistance to endocrine therapy remain elusive. In this study, we elucidate the pivotal role of GPR81, a G protein-coupled receptor, in ER+ breast cancer (BC) by demonstrating low expression of GPR81 in tamoxifen (TAM)-resistant ER+ BC cell lines and tumor samples, along with the underlying molecular mechanisms. MAIN METHODS: Fatty acid oxidation (FAO) levels and lipid accumulation were explored using MDA and FAßO assay, BODIPY 493/503 staining, and Lipid TOX staining. Autophagy levels were assayed using CYTO-ID detection and Western blotting. The impact of GPR81 on TAM resistance in BC was investigated through CCK8 assay, colony formation assay and a xenograft mice model. RESULTS: Aberrantly low GPR81 expression in TAM-resistant BC cells disrupts the Rap1 pathway, leading to the upregulation of PPARα and CPT1. This elevation in PPARα/CPT1 enhances FAO, impedes lipid accumulation and lipid droplet (LD) formation, and subsequently inhibits cell autophagy, ultimately promoting TAM-resistant BC cell growth. Moreover, targeting GPR81 and FAO emerges as a promising therapeutic strategy, as the GPR81 agonist and the CPT1 inhibitor etomoxir effectively inhibit ER+ BC cell and tumor growth in vivo, re-sensitizing TAM-resistant ER+ cells to TAM treatment. CONCLUSION: Our data highlight the critical and functionally significant role of GPR81 in promoting ER+ breast tumorigenesis and resistance to endocrine therapy. GPR81 and FAO levels show potential as diagnostic biomarkers and therapeutic targets in clinical settings for TAM-resistant ER+ BC.


Sujet(s)
Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Acides gras , Souris nude , Oxydoréduction , Récepteur PPAR alpha , Récepteurs couplés aux protéines G , Tamoxifène , Tamoxifène/pharmacologie , Humains , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Femelle , Récepteurs couplés aux protéines G/métabolisme , Animaux , Acides gras/métabolisme , Souris , Récepteur PPAR alpha/métabolisme , Antinéoplasiques hormonaux/pharmacologie , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Autophagie/effets des médicaments et des substances chimiques , Souris de lignée BALB C
12.
Biomed Pharmacother ; 176: 116908, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38850668

RÉSUMÉ

Non-alcoholic fatty liver disease (NAFLD), particularly advanced non-alcoholic steatohepatitis (NASH), leads to irreversible liver damage. This study investigated the therapeutic effects and potential mechanism of a novel extract from traditional Chinese medicine Alisma orientale (Sam.) Juzep (AE) on free fatty acid (FFA)-induced HepG2 cell model and high-fat diet (HFD) + carbon tetrachloride (CCl4)-induced mouse model of NASH. C57BL/6 J mice were fed a HFD for 10 weeks. Subsequently, the mice were injected with CCl4 to induce NASH and simultaneously treated with AE at daily doses of 50, 100, and 200 mg/kg for 4 weeks. At the end of the treatment, animals were fasted for 12 h and then sacrificed. Blood samples and liver tissues were collected for analysis. Lipid profiles, oxidative stress, and histopathology were examined. Additionally, a polymerase chain reaction (PCR) array was used to predict the molecular targets and potential mechanisms involved, which were further validated in vivo and in vitro. The results demonstrated that AE reversed liver damage (plasma levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), hepatocyte ballooning, hepatic steatosis, and NAS score), the accumulation of hepatic lipids (TG and TC), and oxidative stress (MDA and GSH). PCR array analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that AE protects against NASH by regulating the adipocytokine signaling pathway and influencing nuclear receptors such as PPARα. Furthermore, AE increased the expression of peroxisome proliferator-activated receptor gamma coactivator-1α (PPARGC1α) and reversed the decreased expression of PPARα in NASH mice. Moreover, in HepG2 cells, AE reduced FFA-induced lipid accumulation and oxidative stress, which was dependent on PPARα up-regulation. Overall, our findings suggest that AE may serve as a potential therapeutic approach for NASH by inhibiting lipid accumulation and reducing oxidative stress specifically through the PPARα pathway.


Sujet(s)
Alisma , Alimentation riche en graisse , Stéatose hépatique non alcoolique , Extraits de plantes , Transduction du signal , Animaux , Humains , Mâle , Souris , Alisma/composition chimique , Tétrachloro-méthane , Alimentation riche en graisse/effets indésirables , Modèles animaux de maladie humaine , Cellules HepG2 , Métabolisme lipidique/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Souris de lignée C57BL , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Extraits de plantes/isolement et purification , Récepteur PPAR alpha/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
13.
Food Res Int ; 188: 114489, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38823872

RÉSUMÉ

Solanum nigrum L. (SN) berry is an edible berry containing abundant polyphenols and bioactive compounds, which possess antioxidant and antiinflammatory properties. However, the effects of SN on alcohol-induced biochemical changes in the enterohepatic axis remain unclear. In the current study, a chronic ethanol-fed mice ALD model was used to test the protective mechanisms of SN berries. Microbiota composition was determined via 16S rRNA sequencing, we found that SN berries extract (SNE) improved intestinal imbalance by reducing the Firmicutes to Bacteroides ratio, restoring the abundance of Akkermansia microbiota, and reducing the abundance of Allobaculum and Shigella. SNE restored the intestinal short-chain fatty acids content. In addition, liver transcriptome data analysis revealed that SNE primarily affected the genes involved in lipid metabolism and inflammatory responses. Furthermore, SNE ameliorated hepatic steatosis in alcohol-fed mice by activating AMP-activated protein kinase (AMPK), acetyl-CoA carboxylase (ACC), peroxisome proliferator-activated receptor α (PPAR-α). SNE reduced the expression of toll-like receptor 4 (TLR4), myeloid differentiation factor-88 (MyD88) nuclear factor kappa-B (NF-κB), which can indicate that SNE mainly adjusted LPS/TLR4/MyD88/NF-κB pathway to reduce liver inflammation. SNE enhanced hepatic antioxidant capacity by regulating NRF2-related protein expression. SNE alleviates alcoholic liver injury by regulating of gut microbiota, lipid metabolism, inflammation, and oxidative stress. This study may provide a reference for the development and utilization of SN resources.


Sujet(s)
Fruit , Microbiome gastro-intestinal , Métabolisme lipidique , Maladies alcooliques du foie , Stress oxydatif , Extraits de plantes , Solanum nigrum , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Métabolisme lipidique/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Souris , Fruit/composition chimique , Solanum nigrum/composition chimique , Mâle , Maladies alcooliques du foie/métabolisme , Maladies alcooliques du foie/prévention et contrôle , Souris de lignée C57BL , Inflammation , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Récepteur de type Toll-4/métabolisme , Modèles animaux de maladie humaine , Récepteur PPAR alpha/métabolisme , Antioxydants/pharmacologie , Éthanol
14.
J Biol Chem ; 300(6): 107340, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38705390

RÉSUMÉ

Triclosan (TCS) is an antimicrobial toxicant found in a myriad of consumer products and has been detected in human tissues, including breastmilk. We have evaluated the impact of lactational TCS on UDP-glucuronosyltransferase 1A1 (UGT1A1) expression and bilirubin metabolism in humanized UGT1 (hUGT1) neonatal mice. In hUGT1 mice, expression of the hepatic UGT1A1 gene is developmentally delayed resulting in elevated total serum bilirubin (TSB) levels. We found that newborn hUGT1 mice breastfed or orally treated with TCS presented lower TSB levels along with induction of hepatic UGT1A1. Lactational and oral treatment by gavage with TCS leads to the activation of hepatic nuclear receptors constitutive androstane receptor (CAR), peroxisome proliferator-activated receptor alpha (PPARα), and stress sensor, activating transcription factor 4 (ATF4). When CAR-deficient hUGT1 mice (hUGT1/Car-/-) were treated with TCS, TSB levels were reduced with a robust induction of hepatic UGT1A1, leaving us to conclude that CAR is not tied to UGT1A1 induction. Alternatively, when PPARα-deficient hUGT1 mice (hUGT1/Pparα-/-) were treated with TCS, hepatic UGT1A1 was not induced. Additionally, we had previously demonstrated that TCS is a potent inducer of ATF4, a transcriptional factor linked to the integrated stress response. When ATF4 was deleted in liver of hUGT1 mice (hUGT1/Atf4ΔHep) and these mice treated with TCS, we observed superinduction of hepatic UGT1A1. Oxidative stress genes in livers of hUGT1/Atf4ΔHep treated with TCS were increased, suggesting that ATF4 protects liver from excessive oxidative stress. The increase oxidative stress may be associated with superinduction of UGT1A1. The expression of ATF4 in neonatal hUGT1 hepatic tissue may play a role in the developmental repression of UGT1A1.


Sujet(s)
Facteur de transcription ATF-4 , Animaux nouveau-nés , Bilirubine , Glucuronosyltransferase , Foie , Récepteur PPAR alpha , Triclosan , Animaux , Glucuronosyltransferase/métabolisme , Glucuronosyltransferase/génétique , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique , Souris , Facteur de transcription ATF-4/métabolisme , Facteur de transcription ATF-4/génétique , Triclosan/pharmacologie , Humains , Bilirubine/pharmacologie , Bilirubine/métabolisme , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Souris knockout , Femelle , Récepteur constitutif des androstanes , Récepteurs cytoplasmiques et nucléaires/métabolisme , Récepteurs cytoplasmiques et nucléaires/génétique
15.
Mol Metab ; 85: 101958, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38763495

RÉSUMÉ

OBJECTIVE: The prevalence of metabolic diseases is increasing globally at an alarming rate; thus, it is essential that effective, accessible, low-cost therapeutics are developed. Peroxisome proliferator-activated receptors (PPARs) are transcription factors that tightly regulate glucose homeostasis and lipid metabolism and are important drug targets for the treatment of type 2 diabetes and dyslipidemia. We previously identified LDT409, a fatty acid-like compound derived from cashew nut shell liquid, as a novel pan-active PPARα/γ/δ compound. Herein, we aimed to assess the efficacy of LDT409 in vivo and investigate the molecular mechanisms governing the actions of the fatty acid mimetic LDT409 in diet-induced obese mice. METHODS: C57Bl/6 mice (6-11-month-old) were fed a chow or high fat diet (HFD) for 4 weeks; mice thereafter received once daily intraperitoneal injections of vehicle, 10 mg/kg Rosiglitazone, 40 mg/kg WY14643, or 40 mg/kg LDT409 for 18 days while continuing the HFD. During treatments, body weight, food intake, glucose and insulin tolerance, energy expenditure, and intestinal lipid absorption were measured. On day 18 of treatment, tissues and plasma were collected for histological, molecular, and biochemical analysis. RESULTS: We found that treatment with LDT409 was effective at reversing HFD-induced obesity and associated metabolic abnormalities in mice. LDT409 lowered food intake and hyperlipidemia, while improving insulin tolerance. Despite being a substrate of both PPARα and PPARγ, LDT409 was crucial for promoting hepatic fatty acid oxidation and reducing hepatic steatosis in HFD-fed mice. We also highlighted a role for LDT409 in white and brown adipocytes in vitro and in vivo where it decreased fat accumulation, increased lipolysis, induced browning of WAT, and upregulated thermogenic gene Ucp1. Remarkably, LDT409 reversed HFD-induced weight gain back to chow-fed control levels. We determined that the LDT409-induced weight-loss was associated with a combination of increased energy expenditure (detectable before weight loss was apparent), decreased food intake, increased systemic fat utilization, and increased fecal lipid excretion in HFD-fed mice. CONCLUSIONS: Collectively, LDT409 represents a fatty acid mimetic that generates a uniquely favorable metabolic response for the treatment of multiple abnormalities including obesity, dyslipidemia, metabolic dysfunction-associated steatotic liver disease, and diabetes. LDT409 is derived from a highly abundant natural product-based starting material and its development could be pursued as a therapeutic solution to the global metabolic health crisis.


Sujet(s)
Alimentation riche en graisse , Acides gras , Souris de lignée C57BL , Obésité , Animaux , Souris , Obésité/métabolisme , Obésité/traitement médicamenteux , Alimentation riche en graisse/effets indésirables , Mâle , Acides gras/métabolisme , Stéatose hépatique/métabolisme , Stéatose hépatique/traitement médicamenteux , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/agonistes , Métabolisme lipidique/effets des médicaments et des substances chimiques , Récepteurs activés par les proliférateurs de peroxysomes/métabolisme , Récepteurs activés par les proliférateurs de peroxysomes/agonistes , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/étiologie
16.
Obesity (Silver Spring) ; 32(7): 1349-1361, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38816990

RÉSUMÉ

OBJECTIVE: Metabolic dysfunction-associated steatotic liver disease (MASLD) is becoming an escalating health problem in pediatric populations. This study aimed to investigate the role of N-acetyltransferase 10 (NAT10) in maternal high-fat diet (HFD)-induced MASLD in offspring at early life. METHODS: We generated male hepatocyte-specific NAT10 knockout (Nat10HKO) mice and mated them with female Nat10fl/fl mice under chow or HFD feeding. Body weight, liver histopathology, and expression of lipid metabolism-associated genes (Srebp1c, Fasn, Pparα, Cd36, Fatp2, Mttp, and Apob) were assessed in male offspring at weaning. Lipid uptake assays were performed both in vivo and in vitro. The mRNA stability assessment and RNA immunoprecipitation were performed to determine NAT10-regulated target genes. RESULTS: NAT10 deletion in hepatocytes of male offspring alleviated perinatal lipid accumulation induced by maternal HFD, decreasing expression levels of Srebp1c, Fasn, Cd36, Fatp2, Mttp, and Apob while enhancing Pparα expression. Furthermore, Nat10HKO male mice exhibited reduced lipid uptake. In vitro, NAT10 promoted lipid uptake by enhancing the mRNA stability of CD36 and FATP2. RNA immunoprecipitation assays exhibited direct interactions between NAT10 and CD36/FATP2 mRNA. CONCLUSIONS: NAT10 deletion in offspring hepatocytes ameliorates maternal HFD-induced hepatic steatosis through decreasing mRNA stability of CD36 and FATP2, highlighting NAT10 as a potential therapeutic target for pediatric MASLD.


Sujet(s)
Alimentation riche en graisse , Stéatose hépatique , Hépatocytes , Métabolisme lipidique , Foie , Souris knockout , Animaux , Alimentation riche en graisse/effets indésirables , Mâle , Femelle , Souris , Grossesse , Foie/métabolisme , Foie/anatomopathologie , Hépatocytes/métabolisme , Stéatose hépatique/étiologie , Stéatose hépatique/métabolisme , Acetyltransferases/génétique , Acetyltransferases/métabolisme , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Protéine-1 de liaison à l'élément de régulation des stérols/métabolisme , Protéine-1 de liaison à l'élément de régulation des stérols/génétique , Effets différés de l'exposition prénatale à des facteurs de risque , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique , Souris de lignée C57BL , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/étiologie
17.
Phytomedicine ; 130: 155705, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38761776

RÉSUMÉ

BACKGROUND: Senolytic combination of dasatinib and quercetin (DQ) is the most studied senolytics drugs used to treat various age-related diseases. However, its protective activity against diabetic kidney disease (DKD) and underlying mechanisms are uncertain. PURPOSE: To investigate the functions and potential mechanisms of the senolytics DQ on DKD. METHODS: Diabetic db/db mice were administrated DQ or transfected with over-expressed PPARα or shPPARα vector. The positive control group was administered irbesartan. Renal function and fibrotic changes in kidney tissue were tested. Single-cell RNA-seq (scRNA-seq) was conducted to analyze the differential transcriptome between the diabetic and control mice. Molecular docking simulation was used to assess the combination of DQ and potential factors. Moreover, tubular epithelial cells under high-glucose (HG) conditions were incubated with DQ and transfected with or without over-expressed PPARα/siPPARα vector. RESULTS: DQ significantly improved renal function, histopathological and fibrotic changes, alleviated lipid deposition, and increased ATP levels in mice with DKD. DQ reduced multiple fatty acid oxidation (FAO) pathway-related proteins and up-regulated PPARα in db/db mice. Overexpression of PPARα upregulated the expression of PPARα-targeting downstream FAO pathway-related proteins, restored renal function, and inhibited renal fibrosis in vitro and in vivo. Moreover, molecular docking and dynamics simulation analyses indicated the nephroprotective effect of DQ via binding to PPARα. Knockdown of PPARα reversed the effect of DQ on the FAO pathway and impaired the protective effect of DQ during DKD. CONCLUSION: For the first time, DQ was found to exert a renal protective effect by binding to PPARα and attenuating renal damage through the promotion of FAO in DKD.


Sujet(s)
Dasatinib , Néphropathies diabétiques , Simulation de docking moléculaire , Récepteur PPAR alpha , Quercétine , Animaux , Néphropathies diabétiques/traitement médicamenteux , Quercétine/pharmacologie , Récepteur PPAR alpha/métabolisme , Souris , Dasatinib/pharmacologie , Mâle , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Souris de lignée C57BL , Diabète expérimental/traitement médicamenteux , Diabète expérimental/complications
18.
Free Radic Biol Med ; 221: 81-88, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38762061

RÉSUMÉ

Androgen receptor (AR)-targeting therapy induces oxidative stress in prostate cancer. However, the mechanism of oxidative stress induction by AR-targeting therapy remains unclear. This study investigated the mechanism of oxidative stress induction by AR-targeting therapy, with the aim to develop novel therapeutics targeting oxidative stress induced by AR-targeting therapy. Intracellular reactive oxygen species (ROS) was examined by fluorescence microscopy and flow cytometry analysis. The effects of silencing gene expression and small molecule inhibitors on gene expression and cytotoxic effects were examined by quantitative real-time PCR and cell proliferation assay. ROS induced by androgen depletion co-localized with peroxisomes in prostate cancer cells. Among peroxisome-related genes, PPARA was commonly induced by AR inhibition and involved in ROS production via PKC signaling. Inhibition of PPARα by specific siRNA and a small molecule inhibitor suppressed cell proliferation and increased cellular sensitivity to the antiandrogen enzalutamide in prostate cancer cells. This study revealed a novel pathway by which AR inhibition induced intracellular ROS mainly in peroxisomes through PPARα activation in prostate cancer. This pathway is a promising target for the development of novel therapeutics for prostate cancer in combination with AR-targeting therapy such as antiandrogen enzalutamide.


Sujet(s)
Benzamides , Prolifération cellulaire , Résistance aux médicaments antinéoplasiques , Nitriles , Stress oxydatif , Péroxysomes , 3-Phényl-2-thiohydantoïne , Tumeurs de la prostate , Récepteurs aux androgènes , Humains , Mâle , Antagonistes du récepteur des androgènes/pharmacologie , Benzamides/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Nitriles/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Péroxysomes/métabolisme , Péroxysomes/effets des médicaments et des substances chimiques , 3-Phényl-2-thiohydantoïne/pharmacologie , Récepteur PPAR alpha/métabolisme , Récepteur PPAR alpha/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Espèces réactives de l'oxygène/métabolisme , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Petit ARN interférent/génétique , Transduction du signal/effets des médicaments et des substances chimiques
19.
Phytomedicine ; 130: 155774, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38820659

RÉSUMÉ

BACKGROUND: Metabolic and alcohol-associated liver disease (MetALD) shows a high prevalence rate in liver patients, but there is currently no effective treatment for MetALD. As a typical edible traditional Chinese medicinal herb, the anti-inflammatory, antioxidant, and hepatoprotective properties of water extract of Chrysanthemum morifolium Ramat. (WECM) has been demonstrated. However, its therapeutic effect on MetALD and the associated mechanisms remain unclear. PURPOSE: To investigate the underlying mechanisms of WECM against MetALD. METHODS: We constructed a MetALD rat model following a high-fat & high-sucrose plus alcohol diet (HFHSAD). MetALD rats were treated with WECM at 2.1, 4.2, and 8.4 g/kg/d for six weeks. Efficacy was determined, and pathways associated with WECM against MetALD were predicted through serum and hepatic biochemical marker measurement, histopathological section analysis, 16S rDNA sequencing of the gut microbiota and untargeted serum metabolomics analyses. Changes in genes and proteins in the peroxisome proliferator-activated receptor alpha (PPARα) and gamma (PPARγ) signaling pathways were detected by RT‒PCR and Western blotting. RESULTS: WECM treatment significantly attenuated hepatic steatosis, hyperlipidemia and markers of liver injury in MetALD rats. Moreover, WECM improved vascular endothelial function, hypertension, and systematic oxidative stress. Mechanistically, WECM treatment altered the overall structure of the gut microbiota through maintaining Firmicutes/Bacteroidota ratio and reducing harmful bacterial abundances such as Clostridium, Faecalibaculum, and Herminiimonas. Notably, WECM promoted 15-deoxy-△12, 14-prostaglandin J2 (15d-PGJ2) release and further activated the PPARγ to reduce serum TNF-α, IL-1ß, and IL-6 levels. Additionally, WECM upregulated PPARα and downregulated the levels of CD36 and FABP4 to improve lipid metabolism. CONCLUSION: Our findings provide the first evidence that WECM treatment significantly improved hepatic steatosis, oxidative stress and inflammation in MetALD rats by regulating the gut microbiota and activating the 15d-PGJ2/PPARγ and PPARα signaling pathway.


Sujet(s)
Chrysanthemum , Microbiome gastro-intestinal , Maladies alcooliques du foie , Récepteur PPAR alpha , Récepteur PPAR gamma , Rat Sprague-Dawley , Chrysanthemum/composition chimique , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Récepteur PPAR gamma/métabolisme , Récepteur PPAR alpha/métabolisme , Mâle , Maladies alcooliques du foie/traitement médicamenteux , Alimentation riche en graisse/effets indésirables , Rats , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Extraits de plantes/pharmacologie , Modèles animaux de maladie humaine , Transduction du signal/effets des médicaments et des substances chimiques , Médicaments issus de plantes chinoises/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques
20.
Toxicol Lett ; 397: 79-88, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38734220

RÉSUMÉ

The activation of pregnane X receptor (PXR) or peroxisome proliferator-activated receptor α (PPARα) can induce liver enlargement. Recently, we reported that PXR or PPARα activation-induced hepatomegaly depends on yes-associated protein (YAP) signaling and is characterized by hepatocyte hypertrophy around the central vein area and hepatocyte proliferation around the portal vein area. However, it remains unclear whether PXR or PPARα activation-induced hepatomegaly can be reversed after the withdrawal of their agonists. In this study, we investigated the regression of enlarged liver to normal size following the withdrawal of PCN or WY-14643 (typical agonists of mouse PXR or PPARα) in C57BL/6 mice. The immunohistochemistry analysis of CTNNB1 and KI67 showed a reversal of hepatocyte size and a decrease in hepatocyte proliferation after the withdrawal of agonists. In details, the expression of PXR or PPARα downstream proteins (CYP3A11, CYP2B10, ACOX1, and CYP4A) and the expression of proliferation-related proteins (CCNA1, CCND1, and PCNA) returned to the normal levels. Furthermore, YAP and its downstream proteins (CTGF, CYR61, and ANKRD1) also restored to the normal states, which was consistent with the change in liver size. These findings demonstrate the reversibility of PXR or PPARα activation-induced hepatomegaly and provide new data for the safety of PXR and PPARα as drug targets.


Sujet(s)
Prolifération cellulaire , Hépatocytes , Hépatomégalie , Foie , Récepteur PPAR alpha , Récepteur du prégnane X , Pyrimidines , Protéines de signalisation YAP , Animaux , Mâle , Souris , Protéines adaptatrices de la transduction du signal/métabolisme , Aryl hydrocarbon hydroxylases , bêta-Caténine/métabolisme , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP4A/métabolisme , Cytochrome P-450 CYP4A/génétique , Famille-2 de cytochromes P450 , Famille-4 de cytochromes P450/génétique , Famille-4 de cytochromes P450/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Hépatomégalie/induit chimiquement , Hépatomégalie/métabolisme , Hépatomégalie/anatomopathologie , Antigène KI-67/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Protéines membranaires , Souris de lignée C57BL , Phosphoprotéines/métabolisme , Phosphoprotéines/génétique , Récepteur PPAR alpha/agonistes , Récepteur PPAR alpha/métabolisme , Récepteur du prégnane X/métabolisme , Récepteur du prégnane X/génétique , Pyrimidines/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Steroid hydroxylases , Protéines de signalisation YAP/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...