Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.128
Filtrer
3.
Nat Commun ; 15(1): 5582, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961093

RÉSUMÉ

Male infertility is a major public health concern globally with unknown etiology in approximately half of cases. The decline in total sperm count over the past four decades and the parallel increase in childhood obesity may suggest an association between these two conditions. Here, we review the molecular mechanisms through which obesity during childhood and adolescence may impair future testicular function. Several mechanisms occurring in obesity can interfere with the delicate metabolic processes taking place at the testicular level during childhood and adolescence, providing the molecular substrate to hypothesize a causal relationship between childhood obesity and the risk of low sperm counts in adulthood.


Sujet(s)
Cellules de Sertoli , Spermatogonies , Mâle , Humains , Cellules de Sertoli/métabolisme , Enfant , Adolescent , Spermatogonies/métabolisme , Infertilité masculine/métabolisme , Maladies métaboliques/métabolisme , Spermatogenèse , Obésité pédiatrique/métabolisme , Testicule/métabolisme , Testicule/croissance et développement , Animaux , Numération des spermatozoïdes
5.
Clin Sci (Lond) ; 138(13): 777-795, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38860674

RÉSUMÉ

Renal tubular injury is considered as the main pathological feature of acute kidney injury (AKI), and mitochondrial dysfunction in renal tubular cells is implicated in the pathogenesis of AKI. The estrogen-related receptor γ (ERRγ) is a member of orphan nuclear receptors which plays a regulatory role in mitochondrial biosynthesis, energy metabolism and many metabolic pathways. Online datasets showed a dominant expression of ERRγ in renal tubules, but the role of ERRγ in AKI is still unknown. In the present study, we investigated the role of ERRγ in the pathogenesis of AKI and the therapeutic efficacy of ERRγ agonist DY131 in several murine models of AKI. ERRγ expression was reduced in kidneys of AKI patients and AKI murine models along with a negative correlation to the severity of AKI. Consistently, silencing ERRγ in vitro enhanced cisplatin-induced tubular cells apoptosis, while ERRγ overexpression in vivo utilizing hydrodynamic-based tail vein plasmid delivery approach alleviated cisplatin-induced AKI. ERRγ agonist DY131 could enhance the transcriptional activity of ERRγ and ameliorate AKI in various murine models. Moreover, DY131 attenuated the mitochondrial dysfunction of renal tubular cells and metabolic disorders of kidneys in AKI, and promoted the expression of the mitochondrial transcriptional factor A (TFAM). Further investigation showed that TFAM could be a target gene of ERRγ and DY131 might ameliorate AKI by enhancing ERRγ-mediated TFAM expression protecting mitochondria. These findings highlighted the protective effect of DY131 on AKI, thus providing a promising therapeutic strategy for AKI.


Sujet(s)
Atteinte rénale aigüe , Récepteurs des oestrogènes , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/génétique , Animaux , Récepteurs des oestrogènes/métabolisme , Humains , Mâle , Souris , Mitochondries/métabolisme , Souris de lignée C57BL , Maladies métaboliques/métabolisme , Apoptose , Modèles animaux de maladie humaine , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Cisplatine , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique
6.
Curr Med Sci ; 44(3): 463-474, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38900388

RÉSUMÉ

Excessive fat deposition in obese subjects promotes the occurrence of metabolic diseases, such as type 2 diabetes mellitus (T2DM), cardiovascular diseases, and non-alcoholic fatty liver disease (NAFLD). Adipose tissue is not only the main form of energy storage but also an endocrine organ that not only secretes adipocytokines but also releases many extracellular vesicles (EVs) that play a role in the regulation of whole-body metabolism. Exosomes are a subtype of EVs, and accumulating evidence indicates that adipose tissue exosomes (AT Exos) mediate crosstalk between adipose tissue and multiple organs by being transferred to targeted cells or tissues through paracrine or endocrine mechanisms. However, the roles of AT Exos in crosstalk with metabolic organs remain to be fully elucidated. In this review, we summarize the latest research progress on the role of AT Exos in the regulation of metabolic disorders. Moreover, we discuss the potential role of AT Exos as biomarkers in metabolic diseases and their clinical application.


Sujet(s)
Tissu adipeux , Exosomes , Maladies métaboliques , Exosomes/métabolisme , Humains , Tissu adipeux/métabolisme , Maladies métaboliques/métabolisme , Diabète de type 2/métabolisme , Marqueurs biologiques/métabolisme , Animaux , Stéatose hépatique non alcoolique/métabolisme , Obésité/métabolisme , Adipokines/métabolisme , Maladie chronique
8.
Nat Metab ; 6(6): 1178-1196, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38867022

RÉSUMÉ

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


Sujet(s)
Modèles animaux de maladie humaine , Stéatose hépatique non alcoolique , Animaux , Humains , Souris , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/anatomopathologie , Mâle , Foie/métabolisme , Foie/anatomopathologie , Maladies métaboliques/métabolisme , Maladies métaboliques/étiologie , Régime occidental/effets indésirables , Études rétrospectives , Cirrhose du foie/métabolisme , Cirrhose du foie/étiologie
9.
In Vivo ; 38(4): 1917-1926, 2024.
Article de Anglais | MEDLINE | ID: mdl-38936925

RÉSUMÉ

BACKGROUND/AIM: To examine the relationship between the body surface area (BSA) and body composition in patients with metabolic dysfunction-associated steatotic liver disease (MASLD, 2,141 men and 986 women). MATERIALS AND METHODS: BSA and body composition parameters were examined. RESULTS: The median body mass index (BMI) was 25.0 kg/m2 for both men and women (p=0.7754). The median body surface area (BSA) was 1.854 m2 for men and 1.618 m2 for women (p<0.0001). In men, the median fat mass was 17.7 kg, whereas in women, it was 22.1 kg (p<0.0001). Additionally, the median fat-free mass was 55.4 kg in men and 39.3 kg in women (p<0.0001).). In male cases, BSA significantly correlated with fat mass (r=0.82, p<0.0001) and fat-free mass (r=0.95, p<0.0001). In female cases, BSA significantly correlated with fat mass (r=0.87, p<0.0001) and fat-free mass (r=0.94, p<0.0001). CONCLUSION: BSA could be a useful marker for the estimation of body composition in patients with MASLD.


Sujet(s)
Composition corporelle , Indice de masse corporelle , Surface corporelle , Humains , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Adulte , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Stéatose hépatique/complications , Maladies métaboliques/métabolisme , Maladies métaboliques/anatomopathologie , Maladies métaboliques/complications , Maladies métaboliques/étiologie
10.
Clin Transl Sci ; 17(6): e13760, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38847320

RÉSUMÉ

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


Sujet(s)
Adiponectine , Modèles animaux de maladie humaine , Hépatocytes , Stéatose hépatique non alcoolique , Animaux , Adiponectine/métabolisme , Adiponectine/pharmacologie , Adiponectine/déficit , Souris , Humains , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/prévention et contrôle , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/étiologie , Mâle , Souris de lignée C57BL , Transduction du signal/effets des médicaments et des substances chimiques , Alimentation riche en graisse/effets indésirables , Erreurs innées du métabolisme/métabolisme , Erreurs innées du métabolisme/traitement médicamenteux , Erreurs innées du métabolisme/anatomopathologie , Maladies métaboliques/traitement médicamenteux , Maladies métaboliques/métabolisme , Maladies métaboliques/prévention et contrôle , Maladies métaboliques/étiologie , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Stéatose hépatique/prévention et contrôle , Stéatose hépatique/métabolisme , Stéatose hépatique/traitement médicamenteux , Stéatose hépatique/anatomopathologie
12.
Int J Mol Sci ; 25(11)2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38891828

RÉSUMÉ

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


Sujet(s)
Stéatose hépatique non alcoolique , Humains , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/thérapie , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/complications , Stéatose hépatique non alcoolique/étiologie , Maladies métaboliques/métabolisme , Maladies métaboliques/étiologie , Stéatose hépatique/métabolisme , Stéatose hépatique/étiologie , Stéatose hépatique/thérapie , Stéatose hépatique/complications , Animaux
13.
Int J Mol Sci ; 25(11)2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38892029

RÉSUMÉ

Cardiometabolic diseases (CMDs) encompass a range of prevalent, often preventable, non-communicable illnesses, including myocardial infarction, stroke, cardiac insufficiency, arterial hypertension, obesity, type 2 diabetes mellitus, insulin resistance, chronic renal dysfunction, non-alcoholic fatty liver disease, and rare metabolic disorders [...].


Sujet(s)
Maladies cardiovasculaires , Humains , Maladies cardiovasculaires/métabolisme , Maladies cardiovasculaires/étiologie , Maladies métaboliques/métabolisme , Maladies métaboliques/étiologie , Diabète de type 2/métabolisme , Diabète de type 2/étiologie , Animaux , Insulinorésistance , Obésité/métabolisme
16.
Sci Rep ; 14(1): 12663, 2024 06 03.
Article de Anglais | MEDLINE | ID: mdl-38830939

RÉSUMÉ

Patients with metabolic dysfunction-associated fatty liver disease (MAFLD) often present with concomitant metabolic dysregulation and alcohol consumption, potentially leading to distinct clinical outcomes. We analyzed data from 8043 participants with MAFLD in the Thai National Health Examination Survey with linked mortality records. According to the MAFLD criteria, 1432 individuals (17.2%) were categorized as having the diabetes phenotype, 5894 (71.0%) as the overweight/obesity phenotype, and 978 (11.8%) as the lean metabolic phenotype. Over 71,145 person-years, 916 participants died. Using Cox proportional hazard models adjusting for physiological, lifestyle, and comorbid factors, both diabetes (adjusted hazards ratio [aHR] 1.59, 95% CI 1.18-2.13) and lean metabolic phenotypes (aHR 1.28, 95% CI 1.01-1.64) exhibited significantly higher mortality risk compared to the overweight/obesity phenotype. A J-shaped relationship was observed between daily alcohol consumption and the risk of all-cause mortality. Daily alcohol intake exceeding 50 g for women and 60 g for men increased the all-cause mortality risk among MAFLD individuals with the lean metabolic phenotype (aHR 3.39, 95% CI 1.02-11.29). Our study found that metabolic phenotype and alcohol consumption have interactive effects on the risk of all-cause mortality in patients with MAFLD, indicating that evaluating both factors is crucial for determining prognostic outcomes and management strategies.


Sujet(s)
Consommation d'alcool , Phénotype , Humains , Mâle , Femelle , Consommation d'alcool/effets indésirables , Adulte d'âge moyen , Adulte , Facteurs de risque , Études de cohortes , Modèles des risques proportionnels , Obésité/complications , Obésité/mortalité , Obésité/métabolisme , Sujet âgé , Thaïlande/épidémiologie , Maladies métaboliques/mortalité , Maladies métaboliques/métabolisme
17.
Commun Biol ; 7(1): 749, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38902371

RÉSUMÉ

Dietary emulsifiers are linked to various diseases. The recent discovery of the role of gut microbiota-host interactions on health and disease warrants the safety reassessment of dietary emulsifiers through the lens of gut microbiota. Lecithin, sucrose fatty acid esters, carboxymethylcellulose (CMC), and mono- and diglycerides (MDG) emulsifiers are common dietary emulsifiers with high exposure levels in the population. This study demonstrates that sucrose fatty acid esters and carboxymethylcellulose induce hyperglycemia and hyperinsulinemia in a mouse model. Lecithin, sucrose fatty acid esters, and CMC disrupt glucose homeostasis in the in vitro insulin-resistance model. MDG impairs circulating lipid and glucose metabolism. All emulsifiers change the intestinal microbiota diversity and induce gut microbiota dysbiosis. Lecithin, sucrose fatty acid esters, and CMC do not impact mucus-bacterial interactions, whereas MDG tends to cause bacterial encroachment into the inner mucus layer and enhance inflammation potential by raising circulating lipopolysaccharide. Our findings demonstrate the safety concerns associated with using dietary emulsifiers, suggesting that they could lead to metabolic syndromes.


Sujet(s)
Dysbiose , Émulsifiants , Microbiome gastro-intestinal , Maladies métaboliques , Animaux , Dysbiose/induit chimiquement , Dysbiose/microbiologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Souris , Mâle , Maladies métaboliques/induit chimiquement , Maladies métaboliques/microbiologie , Maladies métaboliques/métabolisme , Maladies métaboliques/étiologie , Souris de lignée C57BL , Carboxyméthylcellulose de sodium , Saccharose/effets indésirables , Saccharose/administration et posologie , Saccharose/métabolisme , Insulinorésistance , Lécithines
18.
Drug Des Devel Ther ; 18: 2203-2213, 2024.
Article de Anglais | MEDLINE | ID: mdl-38882047

RÉSUMÉ

Mitochondrial carrier homolog 2 (MTCH2) is a member of the solute carrier 25 family, located on the outer mitochondrial membrane. MTCH2 was first identified in 2000. The development in MTCH2 research is rapidly increasing. The most well-known role of MTCH2 is linking to the pro-apoptosis BID to facilitate mitochondrial apoptosis. Genetic variants in MTCH2 have been investigated for their association with metabolic and neurodegenerative diseases, however, no intervention or therapeutic suggestions were provided. Recent studies revealed the physiological and pathological function of MTCH2 in metabolic diseases, neurodegenerative diseases, cancers, embryonic development and reproduction via regulating mitochondrial apoptosis, metabolic shift between glycolysis and oxidative phosphorylation, mitochondrial fusion/fission, epithelial-mesenchymal transition, etc. This review endeavors to assess a total of 131 published articles to summarise the structure and physiological/pathological role of MTCH2, which has not previously been conducted. This review concludes that MTCH2 plays a crucial role in metabolic diseases, neurodegenerative diseases, cancers, embryonic development and reproduction, and the predominant molecular mechanism is regulation of mitochondrial function. This review gives a comprehensive state of current knowledgement on MTCH2, which will promote the therapeutic research of MTCH2.


Sujet(s)
Développement embryonnaire , Maladies métaboliques , Tumeurs , Maladies neurodégénératives , Reproduction , Humains , Maladies neurodégénératives/métabolisme , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Maladies métaboliques/métabolisme , Animaux , Mitochondries/métabolisme , Protéines de transport de la membrane mitochondriale/métabolisme
19.
Int J Mol Sci ; 25(11)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38891759

RÉSUMÉ

Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as nonalcoholic fatty liver disease, is a steatotic liver disease associated with metabolic syndrome (MetS), especially obesity, hypertension, diabetes, hyperlipidemia, and hypertriglyceridemia. MASLD in 43-44% of patients can progress to metabolic dysfunction-associated steatohepatitis (MASH), and 7-30% of these cases will progress to liver scarring (cirrhosis). To date, the mechanism of MASLD and its progression is not completely understood and there were no therapeutic strategies specifically tailored for MASLD/MASH until March 2024. The conventional antiobesity and antidiabetic pharmacological approaches used to reduce the progression of MASLD demonstrated favorable peripheral outcomes but insignificant effects on liver histology. Alternatively, phyto-synthesized metal-based nanoparticles (MNPs) are now being explored in the treatment of various liver diseases due to their unique bioactivities and reduced bystander effects. Although phytonanotherapy has not been explored in the clinical treatment of MASLD/MASH, MNPs such as gold NPs (AuNPs) and silver NPs (AgNPs) have been reported to improve metabolic processes by reducing blood glucose levels, body fat, and inflammation. Therefore, these actions suggest that MNPs can potentially be used in the treatment of MASLD/MASH and related metabolic diseases. Further studies are warranted to investigate the feasibility and efficacy of phytonanomedicine before clinical application.


Sujet(s)
Stéatose hépatique non alcoolique , Phytothérapie , Humains , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/métabolisme , Phytothérapie/méthodes , Nanoparticules métalliques/composition chimique , Nanoparticules métalliques/usage thérapeutique , Animaux , Syndrome métabolique X/traitement médicamenteux , Syndrome métabolique X/métabolisme , Syndrome métabolique X/complications , Maladies métaboliques/traitement médicamenteux , Maladies métaboliques/étiologie , Maladies métaboliques/métabolisme
20.
Int J Mol Sci ; 25(11)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38891865

RÉSUMÉ

The prevalence of metabolic diseases, including type 2 diabetes and metabolic dysfunction-associated steatotic liver disease (MASLD), is steadily increasing. Although many risk factors, such as obesity, insulin resistance, or hyperlipidemia, as well as several metabolic gene programs that contribute to the development of metabolic diseases are known, the underlying molecular mechanisms of these processes are still not fully understood. In recent years, it has become evident that not only protein-coding genes, but also noncoding genes, including a class of noncoding transcripts referred to as long noncoding RNAs (lncRNAs), play key roles in diet-induced metabolic disorders. Here, we provide an overview of selected lncRNA genes whose direct involvement in the development of diet-induced metabolic dysfunctions has been experimentally demonstrated in suitable in vivo mouse models. We further summarize and discuss the associated molecular modes of action for each lncRNA in the respective metabolic disease context. This overview provides examples of lncRNAs with well-established functions in diet-induced metabolic diseases, highlighting the need for appropriate in vivo models and rigorous molecular analyses to assign clear biological functions to lncRNAs.


Sujet(s)
Maladies métaboliques , ARN long non codant , ARN long non codant/génétique , ARN long non codant/métabolisme , Animaux , Humains , Maladies métaboliques/génétique , Maladies métaboliques/métabolisme , Maladies métaboliques/étiologie , Régime alimentaire/effets indésirables , Modèles animaux de maladie humaine , Régulation de l'expression des gènes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...