Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.772
Filtrer
1.
Redox Biol ; 73: 103219, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38851001

RÉSUMÉ

Radiation causes damage to normal tissues that leads to increased oxidative stress, inflammation, and fibrosis, highlighting the need for the selective radioprotection of healthy tissues without hindering radiotherapy effectiveness in cancer. This study shows that adiponectin, an adipokine secreted by adipocytes, protects normal tissues from radiation damage invitro and invivo. Specifically, adiponectin (APN) reduces chronic oxidative stress and fibrosis in irradiated mice. Importantly, APN also conferred no protection from radiation to prostate cancer cells. Adipose tissue is the primary source of circulating endogenous adiponectin. However, this study shows that adipose tissue is sensitive to radiation exposure exhibiting morphological changes and persistent oxidative damage. In addition, radiation results in a significant and chronic reduction in blood APN levels from adipose tissue in mice and human prostate cancer patients exposed to pelvic irradiation. APN levels negatively correlated with bowel toxicity and overall toxicities associated with radiotherapy in prostate cancer patients. Thus, protecting, or modulating APN signaling may improve outcomes for prostate cancer patients undergoing radiotherapy.


Sujet(s)
Adiponectine , Fibrose , Stress oxydatif , Tumeurs de la prostate , Mâle , Animaux , Tumeurs de la prostate/radiothérapie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Humains , Souris , Stress oxydatif/effets des radiations , Adiponectine/métabolisme , Adiponectine/sang , Lésions radiques/métabolisme , Lésions radiques/anatomopathologie , Tissu adipeux/métabolisme , Tissu adipeux/effets des radiations , Radioprotecteurs/pharmacologie , Radioprotecteurs/usage thérapeutique
2.
BMC Genomics ; 25(1): 592, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38867146

RÉSUMÉ

BACKGROUND: Intramuscular fat content is an important index reflecting the quality of mutton, which directly affects the flavor and tenderness of mutton. Livestock and poultry intramuscular fat content is influenced by genetics, nutritional level, and environmental factors. Key regulatory factors play a crucial role in intramuscular fat deposition. However, there is a limited amount of research on the identification and function of key genes involved in intramuscular fat content deposition specifically in sheep. RESULTS: Histological differences in the longest dorsal muscle of the small-tailed frigid sheep increased in diameter and decreased in several muscle fibers with increasing monthly age; The intramuscular fat content of the longest dorsal muscle of the small-tailed cold sheep varied with age, with a minimum of 1 month of age, a maximum of 6 months of age, and a minimum of 12 months of age. Transcriptomic sequencing and bioinformatics analysis revealed a large number of differential genes in the longest dorsal muscles of little-tailed billy goats of different months of age, which were enriched in multiple GO entries and KEGG pathways. Among them, the pathway associated with intramuscular fat was the AMPK signaling pathway, and the related genes were PPARGC1A and ADIPOQ; Immunohistochemical studies showed that PPARGC1A and ADIPOQ proteins were expressed in connective tissues, cell membranes, and, to a lesser extent, the cytoplasm of the longest dorsal muscle of the little-tailed frigid sheep; Real-time PCR and Western Blot validation showed that PPARGC1A and ADIPOQ were both expressed in the longest dorsal muscle of the little-tailed frigid sheep at different ages, and there were age differences in the amount of expression. The ADIPOQ gene was negatively correlated with the intramuscular fat content of the longest dorsal muscle, and the PPARGC1A gene was positively correlated with the intramuscular fat content of the longest dorsal muscle; As inferred from the above results, the ADIPOQ gene was negatively correlated with the intramuscular fat content of the longest dorsal muscle (r = -0.793, P < 0.05); and the PPARGC1A gene was positively correlated with the intramuscular fat content of the longest dorsal muscle r = 0.923, P < 0.05). CONCLUSIONS: Based on the above results, it can be inferred that the ADIPOQ gene is negatively correlated with the intramuscular fat content of the longest back muscle (r = -0.793, P < 0.05); the PPARGC1A gene is positively correlated with the intramuscular fat content of the longest back muscle (r = 0.923, P < 0.05).


Sujet(s)
Tissu adipeux , Muscles squelettiques , Animaux , Ovis/génétique , Ovis/métabolisme , Muscles squelettiques/métabolisme , Tissu adipeux/métabolisme , Adiponectine/métabolisme , Adiponectine/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 , Analyse de profil d'expression de gènes , Transcriptome
3.
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
4.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 178-186, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38836664

RÉSUMÉ

This study aimed to explore the regulatory effects and associated mechanisms of adiponectin on apoptosis and proliferation in the LN18 glioma cell line through the AMPK and Akt signaling pathways. Additionally, we sought to elucidate the impact of adiponectin on the chemosensitivity of the LN18 glioma cell line to temozolomide (TMZ). The proliferation rate of glioma cells treated with adiponectin was assessed using the cholecystokinin (CCK8) assay. The Western blot analysis was employed to assess the expression of p-Akt, p-AMPK, p-mTOR, cleaved caspase3, Bax, Cyclin D1, and Cyclin B1 following adiponectin treatment. Cell apoptosis was quantified using AnnexinV/PI flow cytometry, while changes in the cell cycle were detected using PI staining flow cytometry. The findings revealed that adiponectin upregulates p-AMPK expression and downregulates p-mTOR expression in the PTEN wild-type glioma cell line LN18, with no discernible effect on p-Akt expression. Moreover, adiponectin inhibits the proliferation rate of the PTEN wild-type glioma cell line LN18, enhances the expression of cleaved caspase3 and Bax, and significantly elevates the apoptosis rate, as evidenced by AnnexinV/PI flow cytometry. Adiponectin was observed to suppress the expression of Cyclin D1 and Cyclin B1, increase the number of cells in the G1 phase, and promote autophagy. Additionally, adiponectin augments the expression of Beclin1 and the ratio of LC3II/I in the PTEN wild-type glioma cell line LN18, while decreasing p62 expression. In conclusion, this study posits that adiponectin holds therapeutic promise for glioma treatment. Furthermore, adiponectin enhances the inhibitory effect of TMZ on the proliferation rate of LN18 cells when treated with 0.1 mM and 1 mM TMZ. These results collectively suggest that adiponectin impedes proliferation, encourages apoptosis and autophagy in the LN18 glioma cell line, and heightens its sensitivity to the chemotherapeutic drug TMZ.


Sujet(s)
Adiponectine , Apoptose , Autophagie , Prolifération cellulaire , Gliome , Témozolomide , Adiponectine/métabolisme , Adiponectine/pharmacologie , Adiponectine/génétique , Apoptose/effets des médicaments et des substances chimiques , Humains , Gliome/anatomopathologie , Gliome/métabolisme , Gliome/traitement médicamenteux , Gliome/génétique , Autophagie/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Témozolomide/pharmacologie , Protéines proto-oncogènes c-akt/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , AMP-Activated Protein Kinases/métabolisme
5.
Philos Trans R Soc Lond B Biol Sci ; 379(1906): 20230221, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-38853554

RÉSUMÉ

Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability and is the leading known single-gene cause of autism spectrum disorder. Patients with FXS display varied behavioural deficits that include mild to severe cognitive impairments in addition to mood disorders. Currently, there is no cure for this condition; however, there is an emerging focus on therapies that inhibit mechanistic target of rapamycin (mTOR)-dependent protein synthesis owing to the clinical effectiveness of metformin for alleviating some behavioural symptoms in FXS. Adiponectin (APN) is a neurohormone that is released by adipocytes and provides an alternative means to inhibit mTOR activation in the brain. In these studies, we show that Fmr1 knockout mice, like patients with FXS, show reduced levels of circulating APN and that both long-term potentiation (LTP) and long-term depression (LTD) in the dentate gyrus (DG) are impaired. Brief (20 min) incubation of hippocampal slices in APN (50 nM) was able to rescue both LTP and LTD in the DG and increased both the surface expression and phosphorylation of GluA1 receptors. These results provide evidence for reduced APN levels in FXS playing a role in decreasing bidirectional synaptic plasticity and show that therapies which enhance APN levels may have therapeutic potential for this and related conditions.This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.


Sujet(s)
Adiponectine , Gyrus denté , Modèles animaux de maladie humaine , Protéine du syndrome X fragile , Syndrome du chromosome X fragile , Souris knockout , Plasticité neuronale , Animaux , Syndrome du chromosome X fragile/physiopathologie , Syndrome du chromosome X fragile/traitement médicamenteux , Syndrome du chromosome X fragile/métabolisme , Gyrus denté/métabolisme , Gyrus denté/effets des médicaments et des substances chimiques , Souris , Plasticité neuronale/effets des médicaments et des substances chimiques , Protéine du syndrome X fragile/métabolisme , Protéine du syndrome X fragile/génétique , Adiponectine/métabolisme , Potentialisation à long terme/effets des médicaments et des substances chimiques , Mâle , Récepteur de l'AMPA/métabolisme
6.
Front Biosci (Landmark Ed) ; 29(6): 236, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38940054

RÉSUMÉ

BACKGROUND: This study aimed to elucidate the molecular mechanism through which C1q/tumor necrosis factor (TNF)-related protein 9 (CTRP9) acts in the formation and differentiation of brown adipose tissue (BAT). METHODS: Adenovirus particles encoding CTRP9 and green fluorescent protein were inoculated into the scapula of C57BL/6J mice and fed a high-fat diet for 8 weeks; the body weight, lipid droplet morphology, glucose tolerance, insulin tolerance, and protein expression levels were analyzed. In addition, CTRP9 adenovirus was transfected into brown preadipocytes, and differentiation was induced to identify the effect of CTRP9 overexpression on adipocyte differentiation. RESULTS: CTRP9 overexpression significantly increased the weight gain of mice. Additionally, the CTRP9 overexpression group exhibited significantly increased adipose tissue weight and glucose clearance rates and decreased insulin sensitivity and serum triglyceride levels compared to the control group. Furthermore, CTRP9 overexpression significantly upregulated the adipose triglyceride lipase (ATGL) and perilipin 1 protein expression levels in BAT. The cell experiment results confirmed that CTRP9 overexpression significantly inhibited the adipogenesis of brown adipocytes as evidenced by the downregulation of uncoupling protein 1, beta-3 adrenergic receptor, ATGL, and hormone-sensitive lipase mRNA levels and the significant suppression of uncoupling protein 1, ATGL, and perilipin 1 protein levels in brown adipocytes. CONCLUSIONS: The finding of this study demonstrated that CTRP9 promotes lipolysis by upregulating ATGL expression in vivo and inhibits the differentiation of brown preadipocytes in vitro.


Sujet(s)
Tissu adipeux brun , Alimentation riche en graisse , Lipolyse , Souris de lignée C57BL , Animaux , Alimentation riche en graisse/effets indésirables , Tissu adipeux brun/métabolisme , Mâle , Souris , Adiponectine/métabolisme , Adiponectine/génétique , Insulinorésistance , Triacylglycerol lipase/métabolisme , Triacylglycerol lipase/génétique , Différenciation cellulaire , Adipogenèse/génétique , Périlipine-1/métabolisme , Périlipine-1/génétique , Acyltransferases , Glycoprotéines
7.
Am J Physiol Regul Integr Comp Physiol ; 327(1): R54-R65, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38738295

RÉSUMÉ

Obesity is a major public health issue due to its association with type 2 diabetes, hypertension, and other cardiovascular risks. The BBSome, a complex of eight conserved Bardet-Biedl syndrome (BBS) proteins, has emerged as a key regulator of energy and glucose homeostasis as well as cardiovascular function. However, the importance of adipocyte BBSome in controlling these physiological processes is not clear. Here, we show that adipocyte-specific constitutive disruption of the BBSome through selective deletion of the Bbs1 gene adiponectin (AdipoCre/Bbs1fl/fl mice) does not affect body weight under normal chow or high-fat and high-sucrose diet (HFHSD). However, constitutive BBSome deficiency caused impairment in glucose tolerance and insulin sensitivity. Similar phenotypes were observed after inducible adipocyte-specific disruption of the BBSome (AdipoCreERT2/Bbs1fl/fl mice). Interestingly, a significant increase in renal sympathetic nerve activity, measured using multifiber recording in the conscious state, was observed in AdipoCre/Bbs1fl/fl mice on both chow and HFHSD. A significant increase in tail-cuff arterial pressure was also observed in chow-fed AdipoCre/Bbs1fl/fl mice, but this was not reproduced when arterial pressure was measured by radiotelemetry. Moreover, AdipoCre/Bbs1fl/fl mice had no significant alterations in vascular reactivity. On the other hand, AdipoCre/Bbs1fl/fl mice displayed impaired baroreceptor reflex sensitivity when fed HFHSD, but not on normal chow. Taken together, these data highlight the relevance of the adipocyte BBSome for the regulation of glucose homeostasis and sympathetic traffic. The BBSome also contributes to baroreflex sensitivity under HFHSD, but not normal chow.NEW & NOTEWORTHY The current study show how genetic manipulation of fat cells impacts various functions of the body including sensitivity to the hormone insulin.


Sujet(s)
Adipocytes , Adiponectine , Animaux , Adipocytes/métabolisme , Adiponectine/métabolisme , Adiponectine/génétique , Souris , Insulinorésistance , Mâle , Obésité/physiopathologie , Obésité/métabolisme , Obésité/génétique , Souris knockout , Système nerveux sympathique/physiopathologie , Alimentation riche en graisse , Souris de lignée C57BL , Modèles animaux de maladie humaine , Maladies du système nerveux autonome/physiopathologie , Maladies du système nerveux autonome/génétique , Maladies du système nerveux autonome/métabolisme , Syndrome de Bardet-Biedl/génétique , Syndrome de Bardet-Biedl/physiopathologie , Syndrome de Bardet-Biedl/métabolisme , Protéines associées aux microtubules
8.
Int J Mol Sci ; 25(10)2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38791193

RÉSUMÉ

Adiponectin is an important adipokine involved in glucose and lipid metabolism, but its secretion and potential role in regulating glucose utilization during ovarian development remains unclear. This study aims to investigate the mechanism and effects of follicle-stimulating hormones (FSHs) on adiponectin secretion and its following impact on glucose transport in the granulosa cells of rat ovaries. A range of experimental techniques were utilized to test our research, including immunoblotting, immunohistochemistry, immunofluorescence, ELISA, histological staining, real-time quantitative PCR, and transcriptome analysis. The immunohistochemistry results indicated that adiponectin was primarily located in the granulosa cells of rat ovaries. In primary granulosa cells cultured in vitro, both Western blot and immunofluorescence assays demonstrated that FSH significantly induced adiponectin secretion within 2 h of incubation, primarily via the PKA signaling pathway rather than the PI3K/AKT pathway. Concurrently, the addition of the AdipoR1/AdipoR2 dual agonist AdipoRon to the culture medium significantly stimulated the protein expression of GLUT1 in rat granulosa cells, resulting in enhanced glucose absorption. Consistent with these in vitro findings, rats injected with eCG (which shares structural and functional similarities with FSH) exhibited significantly increased adiponectin levels in both the ovaries and blood. Moreover, there was a notable elevation in mRNA and protein levels of AdipoRs and GLUTs following eCG administration. Transcriptomic analysis further revealed a positive correlation between the expression of the intraovarian adiponectin system and glucose transporter. The present study represents a novel investigation, demonstrating that FSH stimulates adiponectin secretion in ovarian granulosa cells through the PKA signaling pathway. This mechanism potentially influences glucose transport (GLUT1) and utilization within the ovaries.


Sujet(s)
Adiponectine , Hormone folliculostimulante , Glucose , Cellules de la granulosa , Récepteurs à l'adiponectine , Transduction du signal , Animaux , Femelle , Adiponectine/métabolisme , Adiponectine/génétique , Cellules de la granulosa/métabolisme , Cellules de la granulosa/effets des médicaments et des substances chimiques , Rats , Hormone folliculostimulante/métabolisme , Glucose/métabolisme , Récepteurs à l'adiponectine/métabolisme , Récepteurs à l'adiponectine/génétique , Cellules cultivées , Transporteur de glucose de type 1/métabolisme , Transporteur de glucose de type 1/génétique , Rat Sprague-Dawley , Cyclic AMP-Dependent Protein Kinases/métabolisme , Ovaire/métabolisme , Pipéridines
9.
Biochem Biophys Res Commun ; 717: 150041, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38710142

RÉSUMÉ

Ocular inflammation-associated diseases are leading causes of global visual impairment, with limited treatment options. Adiponectin, a hormone primarily secreted by adipose tissue, binds to its receptors, which are widely distributed throughout the body, exerting powerful physiological regulatory effects. The protective role of adiponectin in various inflammatory diseases has gained increasing attention in recent years. Previous studies have confirmed the presence of adiponectin and its receptors in the eyes. Furthermore, adiponectin and its analogs have shown potential as novel drugs for the treatment of inflammatory eye diseases. This article summarizes the evidence for the interplay between adiponectin and inflammatory eye diseases and provides new perspectives on the diagnostic and therapeutic possibilities of adiponectin.


Sujet(s)
Adiponectine , Inflammation , Récepteurs à l'adiponectine , Transduction du signal , Humains , Adiponectine/métabolisme , Récepteurs à l'adiponectine/métabolisme , Animaux , Inflammation/métabolisme , Maladies de l'oeil/métabolisme , Maladies de l'oeil/traitement médicamenteux
10.
FASEB J ; 38(10): e23669, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38747734

RÉSUMÉ

Amomum xanthioides (AX) has been used as an edible herbal medicine to treat digestive system disorders in Asia. Additionally, Lactobacillus casei is a well-known probiotic commonly used in fermentation processes as a starter. The current study aimed to investigate the potential of Lactobacillus casei-fermented Amomum xanthioides (LAX) in alleviating metabolic disorders induced by high-fat diet (HFD) in a mouse model. LAX significantly reduced the body and fat weight, outperforming AX, yet without suppressing appetite. LAX also markedly ameliorated excessive lipid accumulation and reduced inflammatory cytokine (IL-6) levels in serum superior to AX in association with UCP1 activation and adiponectin elevation. Furthermore, LAX noticeably improved the levels of fasting blood glucose, serum insulin, and HOMA-IR through positive regulation of glucose transporters (GLUT2, GLUT4), and insulin receptor gene expression. In conclusion, the fermentation of AX demonstrates a pronounced mitigation of overnutrition-induced metabolic dysfunction, including hyperlipidemia, hyperglycemia, hyperinsulinemia, and obesity, compared to non-fermented AX. Consequently, we proposed that the fermentation of AX holds promise as a potential candidate for effectively ameliorating metabolic disorders.


Sujet(s)
Amomum , Alimentation riche en graisse , Fermentation , Lacticaseibacillus casei , Obésité , Animaux , Alimentation riche en graisse/effets indésirables , Souris , Obésité/métabolisme , Mâle , Lacticaseibacillus casei/métabolisme , Amomum/composition chimique , Souris de lignée C57BL , Probiotiques/pharmacologie , Protéine-1 de découplage/métabolisme , Insulinorésistance , Souris obèse , Adiponectine/métabolisme , Insuline/métabolisme , Insuline/sang , Glycémie/métabolisme
11.
Nat Commun ; 15(1): 3962, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38730247

RÉSUMÉ

Lanifibranor, a pan-PPAR agonist, improves liver histology in patients with metabolic dysfunction-associated steatohepatitis (MASH), who have poor cardiometabolic health (CMH) and cardiovascular events as major mortality cause. NATIVE trial secondary and exploratory outcomes (ClinicalTrials.gov NCT03008070) were analyzed for the effect of lanifibranor on IR, lipid and glucose metabolism, systemic inflammation, blood pressure (BP), hepatic steatosis (imaging and histological grading) for all patients of the original analysis. With lanifibranor, triglycerides, HDL-C, apolipoproteins, insulin, HOMA-IR, HbA1c, fasting glucose (FG), hs-CRP, ferritin, diastolic BP and steatosis improved significantly, independent of diabetes status: most patients with prediabetes returned to normal FG levels. Significant adiponectin increases correlated with hepatic and CMH marker improvement; patients had an average weight gain of 2.5 kg, with 49% gaining ≥2.5% weight. Therapeutic benefits were similar regardless of weight change. Here, we show that effects of lanifibranor on liver histology in MASH are accompanied with CMH improvement, indicative of potential cardiovascular clinical benefits.


Sujet(s)
Chalcones , Adulte , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Adiponectine/métabolisme , Adiponectine/sang , Glycémie/métabolisme , Glycémie/effets des médicaments et des substances chimiques , Pression sanguine/effets des médicaments et des substances chimiques , Maladies cardiovasculaires/traitement médicamenteux , Chalcones/usage thérapeutique , Chalcones/pharmacologie , Stéatose hépatique/traitement médicamenteux , Stéatose hépatique/métabolisme , Insulinorésistance , Métabolisme lipidique/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Foie/métabolisme , Récepteurs activés par les proliférateurs de peroxysomes/agonistes , Récepteurs activés par les proliférateurs de peroxysomes/métabolisme , Propionates , Triglycéride/sang , Triglycéride/métabolisme
12.
PLoS One ; 19(5): e0292997, 2024.
Article de Anglais | MEDLINE | ID: mdl-38728264

RÉSUMÉ

BACKGROUND: Current research suggests that energy transfer through human milk influences infant nutritional development and initiates metabolic programming, influencing eating patterns into adulthood. To date, this research has predominantly been conducted among women in high income settings and/or among undernourished women. We will investigate the relationship between maternal body composition, metabolic hormones in human milk, and infant satiety to explore mechanisms of developmental satiety programming and implications for early infant growth and body composition in Samoans; a population at high risk and prevalence for overweight and obesity. Our aims are (1) to examine how maternal body composition influences metabolic hormone transfer from mother to infant through human milk, and (2) to examine the influences of maternal metabolic hormone transfer and infant feeding patterns on early infant growth and satiety. METHODS: We will examine temporal changes in hormone transfers to infants through human milk in a prospective longitudinal cohort of n = 80 Samoan mother-infant dyads. Data will be collected at three time points (1, 3, & 4 months postpartum). At each study visit we will collect human milk and fingerpick blood samples from breastfeeding mother-infant dyads to measure the hormones leptin, ghrelin, and adiponectin. Additionally, we will obtain body composition measurements from the dyad, observe breastfeeding behavior, conduct semi-structured interviews, and use questionnaires to document infant hunger and feeding cues and satiety responsiveness. Descriptive statistics, univariate and multivariate analyses will be conducted to address each aim. DISCUSSION: This research is designed to advance our understanding of variation in the developmental programming of satiety and implications for early infant growth and body composition. The use of a prospective longitudinal cohort alongside data collection that utilizes a mixed methods approach will allow us to capture a more accurate representation on both biological and cultural variables at play in a population at high risk of overweight and obesity.


Sujet(s)
Composition corporelle , Lait humain , Humains , Lait humain/métabolisme , Lait humain/composition chimique , Femelle , Nourrisson , Études prospectives , Études longitudinales , Leptine/sang , Leptine/métabolisme , Adiponectine/sang , Adiponectine/métabolisme , Adulte , Ghréline/sang , Ghréline/métabolisme , Développement de l'enfant/physiologie , Mâle , Allaitement naturel , Phénomènes physiologiques nutritionnels chez le nourrisson , Satiété/physiologie , Mères
13.
Biofabrication ; 16(3)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38739412

RÉSUMÉ

Reconstruction of large 3D tissues based on assembly of micro-sized multi-cellular spheroids has gained attention in tissue engineering. However, formation of 3D adipose tissue from spheroids has been challenging due to the limited adhesion capability and restricted cell mobility of adipocytes in culture media. In this study, we addressed this problem by developing adipo-inductive nanofibers enabling dual delivery of indomethacin and insulin. These nanofibers were introduced into composite spheroids comprising human adipose-derived stem cells (hADSCs). This approach led to a significant enhancement in the formation of uniform lipid droplets, as evidenced by the significantly increased Oil red O-stained area in spheroids incorporating indomethacin and insulin dual delivery nanofibers (56.9 ± 4.6%) compared to the control (15.6 ± 3.5%) with significantly greater gene expression associated with adipogenesis (C/EBPA, PPARG, FABP4, and adiponectin) of hADSCs. Furthermore, we investigated the influence of culture media on the migration and merging of spheroids and observed significant decrease in migration and merging of spheroids in adipogenic differentiation media. Conversely, the presence of adipo-inductive nanofibers promoted spheroid fusion, allowing the formation of macroscopic 3D adipose tissue in the absence of adipogenic supplements while facilitating homogeneous adipogenesis of hADSCs. The approach described here holds promise for the generation of 3D adipose tissue constructs by scaffold-free assembly of stem cell spheroids with potential applications in clinical and organ models.


Sujet(s)
Adipogenèse , Tissu adipeux , Nanofibres , Sphéroïdes de cellules , Cellules souches , Ingénierie tissulaire , Nanofibres/composition chimique , Humains , Sphéroïdes de cellules/cytologie , Sphéroïdes de cellules/métabolisme , Tissu adipeux/cytologie , Tissu adipeux/métabolisme , Cellules souches/cytologie , Cellules souches/métabolisme , Insuline/métabolisme , Indométacine/pharmacologie , Adipocytes/cytologie , Adipocytes/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Structures d'échafaudage tissulaires/composition chimique , Adiponectine/métabolisme , Cellules cultivées
14.
FASEB J ; 38(10): e23684, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38795334

RÉSUMÉ

Exposure to chronic psychosocial stress is a risk factor for metabolic disorders. Because dipeptidyl peptidase-4 (DPP4) and cysteinyl cathepsin K (CTSK) play important roles in human pathobiology, we investigated the role(s) of DPP4 in stress-related adipocyte differentiation, with a focus on the glucagon-like peptide-1 (GLP-1)/adiponectin-CTSK axis in vivo and in vitro. Plasma and inguinal adipose tissue from non-stress wild-type (DPP4+/+), DPP4-knockout (DPP4-/-) and CTSK-knockout (CTSK-/-) mice, and stressed DPP4+/+, DPP4-/-, CTSK-/-, and DPP4+/+ mice underwent stress exposure plus GLP-1 receptor agonist exenatide loading for 2 weeks and then were analyzed for stress-related biological and/or morphological alterations. On day 14 under chronic stress, stress decreased the weights of adipose tissue and resulted in harmful changes in the plasma levels of DPP4, GLP-1, CTSK, adiponectin, and tumor necrosis factor-α proteins and the adipose tissue levels of CTSK, preadipocyte factor-1, fatty acid binding protein-4, CCAAT/enhancer binding protein-α, GLP-1 receptor, peroxisome proliferator-activated receptor-γ, perilipin2, secreted frizzled-related protein-4, Wnt5α, Wnt11 and ß-catenin proteins and/or mRNAs as well as macrophage infiltration in adipose tissue; these changes were rectified by DPP4 deletion. GLP-1 receptor activation and CTSK deletion mimic the adipose benefits of DPP4 deficiency. In vitro, CTSK silencing and overexpression respectively prevented and facilitated stress serum and oxidative stress-induced adipocyte differentiation accompanied with changes in the levels of pref-1, C/EBP-α, and PPAR-γ in 3T3-L1 cells. Thus, these findings indicated that increased DPP4 plays an essential role in stress-related adipocyte differentiation, possibly through a negative regulation of GLP-1/adiponectin-CTSK axis activation in mice under chronic stress conditions.


Sujet(s)
Adipocytes , Adiponectine , Cathepsine K , Différenciation cellulaire , Dipeptidyl peptidase 4 , Glucagon-like peptide 1 , Souris knockout , Animaux , Souris , Adiponectine/métabolisme , Glucagon-like peptide 1/métabolisme , Adipocytes/métabolisme , Dipeptidyl peptidase 4/métabolisme , Dipeptidyl peptidase 4/génétique , Cathepsine K/métabolisme , Cathepsine K/génétique , Mâle , Souris de lignée C57BL , Stress psychologique/métabolisme , Cellules 3T3-L1 , Exénatide/pharmacologie , Récepteur PPAR gamma/métabolisme , Adipogenèse
15.
Int J Mol Sci ; 25(10)2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38791438

RÉSUMÉ

Geese are susceptible to oxidative stress during reproduction, which can lead to follicular atresia and impact egg production. Follicular atresia is directly triggered by the apoptosis and autophagy of granulosa cells (GCs). Adiponectin (ADPN), which is secreted by adipose tissue, has good antioxidant and anti-apoptotic capacity, but its role in regulating the apoptosis of GCs in geese is unclear. To investigate this, this study examined the levels of oxidative stress, apoptosis, and autophagy in follicular tissues and GCs using RT-qPCR, Western blotting, immunofluorescence, flow cytometry, transcriptomics and other methods. Atretic follicles exhibited high levels of oxidative stress and apoptosis, and autophagic flux was obstructed. Stimulating GCs with H2O2 produced results similar to those of atretic follicles. The effects of ADPN overexpression and knockdown on oxidative stress, apoptosis and autophagy in GCs were investigated. ADPN was found to modulate autophagy and reduced oxidative stress and apoptosis in GCs, in addition to protecting them from H2O2-induced damage. These results may provide a reasonable reference for improving egg-laying performance of geese.


Sujet(s)
Adiponectine , Apoptose , Autophagie , Protéines aviaires , Atrésie folliculaire , Oies , Cellules de la granulosa , Animaux , Femelle , Adiponectine/métabolisme , Adiponectine/génétique , Atrésie folliculaire/métabolisme , Cellules de la granulosa/métabolisme , Peroxyde d'hydrogène/métabolisme , Peroxyde d'hydrogène/pharmacologie , Follicule ovarique/métabolisme , Stress oxydatif , Protéines aviaires/métabolisme
16.
Pol J Pathol ; 75(1): 40-53, 2024.
Article de Anglais | MEDLINE | ID: mdl-38741428

RÉSUMÉ

C1q/TNF-related protein-9 (CTRP9) has been reported to play roles in several types of retinal diseases. However, the role and the potential mechanism of CTRP9 in glaucoma are still incompletely understood. The expression of CTRP9 in OGD/R-induced retinal ganglion cells (RGCs) was detected by quantitative real-time polymerase chain reaction and western blot assay. Cell proliferation was identified by cell counting Kit-8 assay. Flow cytometry, enzyme-linked immunosorbent assay and western blot assay were performed to assess cell apoptosis. Unfolded protein response (UPR), endoplasmic reticulum (ER) stress and the AMPK pathway were evaluated by western blot assay. The data showed that the expression of CTRP9 was significantly downregulated in OGD/R-induced 661W cells. OGD/R treatment reduced cell viability, promoted cell apoptosis and activated the UPR and ER stress. The overexpression of CTRP9 reversed the effects of OGD/R on 661W cell viability, apoptosis, the UPR and ER stress, as well as the AMPK pathway. However, Compound C, an inhibitor of AMPK signaling, reversed the protection of CTRP9 overexpression against injury from OGD/R in 661W cells. In summary, the results revealed that CTRP9 abated the apoptosis and UPR of OGD/R-induced RGCs by regulating the AMPK pathway, which may provide a promising target for the treatment of glaucoma.


Sujet(s)
AMP-Activated Protein Kinases , Apoptose , Stress du réticulum endoplasmique , Cellules ganglionnaires rétiniennes , Transduction du signal , Réponse aux protéines mal repliées , Cellules ganglionnaires rétiniennes/anatomopathologie , Cellules ganglionnaires rétiniennes/métabolisme , Animaux , AMP-Activated Protein Kinases/métabolisme , Souris , Lignée cellulaire , Adiponectine/métabolisme , Survie cellulaire , Glucose/métabolisme , Glaucome/métabolisme , Glaucome/anatomopathologie , Glycoprotéines
17.
Arch Pharm (Weinheim) ; 357(4): e2300631, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38574101

RÉSUMÉ

Nonalcoholic fatty liver disease (NAFLD) is a common liver disorder affecting a quarter of the global residents. Progression of NAFL into nonalcoholic steatohepatitis (NASH) may cause cirrhosis, liver cancer, and failure. Gut microbiota imbalance causes microbial components translocation into the circulation, triggering liver inflammation and NASH-related fibrosis. MicroRNAs (miRNAs) regulate gene expression via repressing target genes. Exosomal miRNAs are diagnostic and prognostic biomarkers for NAFL and NASH liver damage. Our work investigated the role of the gut microbiota in NAFLD pathogenesis via the lipopolysaccharide/toll-like receptor 4/Forkhead box protein O3 (LPS/TLR-4/FoxO3) pathway and certain miRNAs as noninvasive biomarkers for NAFL or its development to NASH. miRNA expression levels were measured using quantitative reverse transcription polymerase chain reaction (qRT-PCR) in 50 NAFL patients, 50 NASH patients, and 50 normal controls. Plasma LPS, TLR-4, adiponectin, peroxisome proliferator-activated receptor γ (PPAR-γ), and FoxO3 concentrations were measured using enzyme-linked immunosorbent assay (ELISA). In NAFL and NASH patients, miR-122, miR-128, FoxO3, TLR-4, LPS, and PPAR-γ were upregulated while miR-200, miR-298, miR-342, and adiponectin were downregulated compared with the normal control. The examined miRNAs might distinguish NAFL and NASH patients from the normal control using receiver operating characteristic analysis. Our study is the first to examine these miRNAs in NAFLD. Our findings imply that these are potentially promising biomarkers for noninvasive early NAFL diagnosis and NASH progression. Understanding the LPS/TLR-4/FoxO3 pathway involvement in NAFL/NASH pathogenesis may aid disease management.


Sujet(s)
microARN , Stéatose hépatique non alcoolique , Humains , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Lipopolysaccharides/pharmacologie , Adiponectine/métabolisme , Protéine O3 à motif en tête de fourche/génétique , Protéine O3 à motif en tête de fourche/métabolisme , Récepteurs activés par les proliférateurs de peroxysomes/métabolisme , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/métabolisme , Relation structure-activité , microARN/génétique , microARN/métabolisme , Marqueurs biologiques/métabolisme , Foie/métabolisme
18.
Poult Sci ; 103(6): 103734, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38636201

RÉSUMÉ

Dietary supplementation with bioactive substances that can regulate lipid metabolism is an effective approach for reducing excessive fat deposition in chickens. Genistein (GEN) has the potential to alleviate fat deposition; however, the underlying mechanism of GEN's fat-reduction action in chickens remains unclear. Therefore, the present study aimed to explore the underlying mechanism of GEN on the reduction of fat deposition from a novel perspective: intercellular transmission of adipokine between adipocytes and hepatocytes. The findings showed that GEN enhanced the secretion of adiponectin (APN) in chicken adipocytes, and the enhancement effect of GEN was completely blocked when the cells were pretreated with inhibitors targeting estrogen receptor ß (ERß) or proliferator-activated receptor γ (PPARγ) signals, respectively. Furthermore, the results demonstrated that both co-treatment with GEN and APN or treatment with the medium supernatant (Med SUP) derived from chicken adipocytes treated with GEN significantly decreased the content of triglyceride and increased the protein levels of ERß, Sirtuin 1 (SIRT1) and phosphor-AMP-activated protein kinase (p-AMPK) in chicken hepatocytes compared to the cells treated with GEN or APN alone. Moreover, the increase in the protein levels of SIRT1 and p-AMPK induced by GEN and APN co-treatment or Med SUP treatment were blocked in chicken hepatocytes pretreated with the inhibitor of ERß signals. Importantly, the up-regulatory effect of GEN and APN co-treatment or Med SUP treatment on the protein level of p-AMPK was also blocked in chicken hepatocytes pretreated with a SIRT1 inhibitor; however, the increase in the protein level of SIRT1 induced by GEN and APN co-treatment or Med SUP treatment was not reversed when the hepatocytes were pretreated with an AMPK inhibitor. In conclusion, the present study demonstrated that GEN enhanced APN secretion by activating the ERß-Erk-PPARγ signaling pathway in chicken adipocytes. Subsequently, adipocyte-derived APN synergized with GEN to activate the ERß-mediated SIRT1-AMPK signaling pathway in chicken hepatocytes, ultimately reducing fat deposition. These findings provide substantial evidence from a novel perspective, supporting the potential use of GEN as a dietary supplement to prevent excessive fat deposition in poultry.


Sujet(s)
Adiponectine , Poulets , Récepteur bêta des oestrogènes , Génistéine , Hépatocytes , Transduction du signal , Sirtuine-1 , Animaux , Génistéine/pharmacologie , Génistéine/administration et posologie , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Sirtuine-1/métabolisme , Récepteur bêta des oestrogènes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Adiponectine/métabolisme , AMP-Activated Protein Kinases/métabolisme , Protéines aviaires/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Tissu adipeux/effets des médicaments et des substances chimiques
19.
Genes (Basel) ; 15(4)2024 04 11.
Article de Anglais | MEDLINE | ID: mdl-38674417

RÉSUMÉ

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a common monogenic disorder characterized by renal cysts and progressive renal failure. In kidney diseases, adipose tissue undergoes functional changes that have been associated with increased inflammation and insulin resistance mediated by release of adipokines. Adiponectin is involved in various cellular processes, such as energy and inflammatory and oxidative processes. However, it remains to be determined whether adiponectin is involved in the concomitant metabolic dysfunctions present in PKD. In this scenario, we aimed to analyze: (a) PPARγ, ADIPOQ, ADIPOR1 and ADIPOR2 gene variations in 92 ADPKD patients through PCR-Sanger sequencing; and (b) adiponectin levels and its oligomerization state by ELISA and Western Blot. Our results indicated that: (a) 14 patients carried the PPARγ SNP, 29 patients carried the ADIPOQ SNP rs1501299, and 25 patients carried the analyzed ADIPOR1 SNPs. Finally, 82 patients carried ADIPOR2 SNPs; and (b) Adiponectin is statistically lower in ADPKD patients compared to controls, and further statistically lower in ESRD than in non-ESRD patients. An inverse relationship between adiponectin and albumin and between adiponectin and creatinine and a direct relationship between adiponectin and eGFR were found. Interestingly, significantly lower levels of adiponectin were found in patients bearing the ADIPOQ rs1501299 SNP and associated with low levels of eGFR. In conclusion, adiponectin levels and the presence of ADIPOQ rs1501299 genotype are significantly associated with a worse ADPKD phenotype, indicating that both could potentially provide important insights into the disease. Further studies are warranted to understand the pathophysiological role of adiponectin in ADPKD patients.


Sujet(s)
Adiponectine , Polykystose rénale autosomique dominante , Polymorphisme de nucléotide simple , Récepteurs à l'adiponectine , Humains , Adiponectine/génétique , Adiponectine/métabolisme , Polykystose rénale autosomique dominante/génétique , Polykystose rénale autosomique dominante/anatomopathologie , Polykystose rénale autosomique dominante/métabolisme , Femelle , Mâle , Récepteurs à l'adiponectine/génétique , Adulte d'âge moyen , Adulte , Récepteur PPAR gamma/génétique , Récepteur PPAR gamma/métabolisme
20.
Obesity (Silver Spring) ; 32(6): 1136-1143, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38644654

RÉSUMÉ

OBJECTIVE: Maternal obesity affects 39.7% of reproductive-age women in the United States. Emerging research has suggested that in utero exposure to maternal obesity is associated with adverse neurodevelopmental outcomes, but knowledge of underlying mechanisms in human samples is lacking. METHODS: A matched case-control study was performed in women with singleton fetuses who were undergoing elective pregnancy termination at gestational ages 15 to 21 weeks. Maternal adiponectin levels from plasma were measured using ELISA kits. RNA was extracted from fetal brain tissue using RNeasy Mini Kit (QIAGEN). mRNA expression from ADIPOR1, ADIPOR2, MTOR, ATG5, ATG7, BECN1, and MAP1LC3B was quantified through the ΔΔCt method and using GAPDH as a housekeeping gene. RESULTS: We have identified transcription patterns associated with inhibition of autophagy in male fetal brain tissue exposed to maternal obesity (↑MTOR, ↓ATG5, ↓ATG7, and ↓MAP1LC3B), with female fetuses demonstrating either no change in transcription or nonsignificant changes associated with increased autophagy. There was significant downregulation of the autophagy-associated gene BECN1 in both male and female individuals who were exposed to obesity in utero. CONCLUSIONS: We present novel evidence suggesting that in utero exposure to maternal obesity in humans may significantly affect neurodevelopment, especially in male fetuses, through alterations in normal autophagy molecular mechanisms and with adiponectin as a potential mediator.


Sujet(s)
Adiponectine , Autophagie , Bécline-1 , Encéphale , Protéines associées aux microtubules , Obésité maternelle , Sérine-thréonine kinases TOR , Humains , Femelle , Grossesse , Mâle , Études cas-témoins , Obésité maternelle/métabolisme , Encéphale/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Adiponectine/métabolisme , Adiponectine/sang , Bécline-1/métabolisme , Adulte , Protéines associées aux microtubules/métabolisme , Protéine-5 associée à l'autophagie/métabolisme , Protéine-5 associée à l'autophagie/génétique , Protéine-7 associée à l'autophagie/génétique , Protéine-7 associée à l'autophagie/métabolisme , Récepteurs à l'adiponectine/métabolisme , Récepteurs à l'adiponectine/génétique , Foetus/métabolisme , ARN messager/métabolisme , Facteurs sexuels , Âge gestationnel , Régulation négative , Obésité/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...