Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.056
Filtrer
1.
Mol Med ; 30(1): 124, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138413

RÉSUMÉ

BACKGROUND: Obesity is well-established as a significant contributor to the development of insulin resistance (IR) and diabetes, partially due to elevated plasma saturated free fatty acids like palmitic acid (PA). Grb10-interacting GYF Protein 2 (GIGYF2), an RNA-binding protein, is widely expressed in various tissues including the liver, and has been implicated in diabetes-induced cognitive impairment. Whereas, its role in obesity-related IR remains uninvestigated. METHODS: In this study, we employed palmitic acid (PA) exposure to establish an in vitro IR model in the human liver cancer cell line HepG2 with high-dose chronic PA treatment. The cells were stained with fluorescent dye 2-NBDG to evaluate cell glucose uptake. The mRNA expression levels of genes were determined by real-time qRT-PCR (RT-qPCR). Western blotting was employed to examine the protein expression levels. The RNA immunoprecipitation (RIP) was used to investigate the binding between protein and mRNA. Lentivirus-mediated gene knockdown and overexpression were employed for gene manipulation. In mice, an IR model induced by a high-fat diet (HFD) was established to validate the role and action mechanisms of GIGYF2 in the modulation of HFD-induced IR in vivo. RESULTS: In hepatocytes, high levels of PA exposure strongly trigger the occurrence of hepatic IR evidenced by reduced glucose uptake and elevated extracellular glucose content, which is remarkably accompanied by up-regulation of GIGYF2. Silencing GIGYF2 ameliorated PA-induced IR and enhanced glucose uptake. Conversely, GIGYF2 overexpression promoted IR, PTEN upregulation, and AKT inactivation. Additionally, PA-induced hepatic IR caused a notable increase in STAU1, which was prevented by depleting GIGYF2. Notably, silencing STAU1 prevented GIGYF2-induced PTEN upregulation, PI3K/AKT pathway inactivation, and IR. STAU1 was found to stabilize PTEN mRNA by binding to its 3'UTR. In liver cells, tocopherol treatment inhibits GIGYF2 expression and mitigates PA-induced IR. In the in vivo mice model, GIGYF2 knockdown and tocopherol administration alleviate high-fat diet (HFD)-induced glucose intolerance and IR, along with the suppression of STAU1/PTEN and restoration of PI3K/AKT signaling. CONCLUSIONS: Our study discloses that GIGYF2 mediates obesity-related IR by disrupting the PI3K/AKT signaling axis through the up-regulation of STAU1/PTEN. Targeting GIGYF2 may offer a potential strategy for treating obesity-related metabolic diseases, including type 2 diabetes.


Sujet(s)
Protéines de transport , Insulinorésistance , Foie , Phosphohydrolase PTEN , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Protéines de liaison à l'ARN , Transduction du signal , Humains , Protéines proto-oncogènes c-akt/métabolisme , Animaux , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Phosphatidylinositol 3-kinases/métabolisme , Souris , Foie/métabolisme , Protéines de transport/métabolisme , Protéines de transport/génétique , Cellules HepG2 , Acide palmitique , Mâle , Protéines du cytosquelette/métabolisme , Protéines du cytosquelette/génétique , Souris de lignée C57BL , Alimentation riche en graisse/effets indésirables
2.
Article de Anglais | MEDLINE | ID: mdl-39147447

RÉSUMÉ

Obesity is a well-known risk factor for testicular function; however, dulaglutide's effect on the testis in obesity has received little attention. Currently, clinicians prescribe the antidiabetic drug dulaglutide only off-label for weight management in non-diabetics. Investigating the impact of this novel compound on obesity is critical for determining whether it has any disruptive effects on testicular cells. We used a well-known animal model of high-fat diet-induced obesity in this investigation, and testicular dysfunction was determined by sperm DNA damage, spermatocyte chromosomal abnormalities, and spermiogram analysis. Following a 12-week high-fat diet challenge, mice were randomly assigned to dulaglutide (0.6 mg/kg/day) or saline treatments for five weeks. Testes and sperm cells were collected 24 h after the last dulaglutide injection. Untreated obese mice had a lower testes/body weight ratio, more sperm DNA damage, diakinesis-metaphase I chromosomal abnormalities, a lower sperm count/motility, more cell morphological defects, and an altered testicular redox balance. In obese mice, dulaglutide injection efficiently restored all disturbed parameters to their control levels. Dulaglutide injection into healthy mice exhibited no significant harmful effects at the applied regimen. As a result, we infer that dulaglutide therapy might bring obese men additional benefits by recovering testicular dysfunction induced by obesity.


Sujet(s)
Alimentation riche en graisse , Modèles animaux de maladie humaine , Peptides glucagon-like , Fragments Fc des immunoglobulines , Obésité , Protéines de fusion recombinantes , Testicule , Animaux , Mâle , Fragments Fc des immunoglobulines/pharmacologie , Obésité/traitement médicamenteux , Peptides glucagon-like/analogues et dérivés , Peptides glucagon-like/pharmacologie , Peptides glucagon-like/usage thérapeutique , Alimentation riche en graisse/effets indésirables , Souris , Protéines de fusion recombinantes/pharmacologie , Testicule/effets des médicaments et des substances chimiques , Testicule/anatomopathologie , Testicule/métabolisme , Altération de l'ADN/effets des médicaments et des substances chimiques , Spermatozoïdes/effets des médicaments et des substances chimiques , Hypoglycémiants/pharmacologie , Mobilité des spermatozoïdes/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Aberrations des chromosomes/effets des médicaments et des substances chimiques , Maladies testiculaires/traitement médicamenteux
3.
J Biochem Mol Toxicol ; 38(9): e23809, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39148263

RÉSUMÉ

Nonalcoholic fatty liver disease (NAFLD) is an alarming ailment that leads to severe liver damage and increases the risk of serious health conditions. The prevalence of NAFLD due to oxidative stress could be mitigated by plant-derived antioxidants. This study aims to investigate the effects of syringic acid (SA) on NAFLD in a high-fat diet (HFD) rat model. Twenty-four rats were randomly divided into four groups (n = 6): normal control, HFD, SA-administered HFD, and positive control SA on a normal diet. Rats in the normal control and positive control groups received a normal diet, and the remaining groups received an HFD for 8 weeks. SA (20 mg/kg b.w.) was orally (gavage) administered for 8 weeks. Lipid profiles were controlled by SA against HFD-fed rats (p < 0.05). SA reduced the serum aspartate aminotransferase and alanine aminotransferase levels by 70%-190%. SA also suppressed pro-inflammatory cytokines and attenuated histopathological and immunohistochemical changes against HFD-fed rats. SA reversed oxidative stress by suppressing the malondialdehyde formation by 82% and replenished the nonenzymatic and enzymatic antioxidant activities (p < 0.05). Gene expressions of nuclear factor-erythroid 2-related factor/heme oxygenase 1 (Nrf2/HO-1) were elevated in SA-treated rats. Ameliorative effects of SA on NAFLD induced by an HFD in rats were prominent through the reversal of oxidative stress and inflammation, regulated by an intrinsic mechanism of defense against oxidative stress, the Nrf2/HO-1 pathway.


Sujet(s)
Acide gallique , Heme oxygenase (decyclizing) , Facteur-2 apparenté à NF-E2 , Stéatose hépatique non alcoolique , Transduction du signal , Animaux , Facteur-2 apparenté à NF-E2/métabolisme , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/prévention et contrôle , Rats , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Acide gallique/analogues et dérivés , Acide gallique/pharmacologie , Heme oxygenase (decyclizing)/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Heme oxygenase-1/métabolisme , Alimentation riche en graisse/effets indésirables , Rat Sprague-Dawley , Antioxydants/pharmacologie , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie
4.
Lipids Health Dis ; 23(1): 250, 2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39154177

RÉSUMÉ

INTRODUCTION: Hypercholesterolemia is associated with increased inflammation and impaired serotonin neurotransmission, potentially contributing to depressive symptoms. However, the role of statins, particularly pitavastatin, in modulating serotonin transporter (SERT) function within this context remains underexplored. This study aimed to investigate whether pitavastatin counteracts the neurobiological effects of hypercholesterolemia. METHODS: Low-density lipoprotein receptor knockout (LDLR-/-) mice on a C57BL/6 background were assigned to three groups: a control group fed a standard chow diet, a group fed a high-fat diet (HFD), and a third group fed a high-fat diet supplemented with pitavastatin (HFD + Pita). We evaluated the effects of HFD with or without pitavastatin on lipid profiles, inflammatory markers, and SERT availability using small-animal positron emission tomography (PET) scans with the radioligand 4-[18F]-ADAM over a 20-week period. RESULTS: Pitavastatin treatment in HFD-fed mice significantly reduced both total cholesterol and LDL cholesterol levels in HFD-fed mice compared to those on HFD alone. Elevated inflammatory markers such as IL-1α, MCP-1/CCL2, and TNF-α in HFD mice were notably decreased in the HFD + Pita group. PET scans showed reduced SERT availability in the brains of HFD mice; however, pitavastatin improved this in brain regions associated with mood regulation, suggesting enhanced serotonin neurotransmission. Additionally, the sucrose preference test showed a trend towards increased preference in the HFD + Pita group compared to the HFD group, indicating a potential reduction in depressive-like behavior. CONCLUSION: Our findings demonstrate that pitavastatin not only lowers cholesterol and reduces inflammation but also enhances SERT availability, suggesting a potential role in alleviating depressive symptoms associated with hypercholesterolemia. These results highlight the multifaceted benefits of pitavastatin, extending beyond its lipid-lowering effects to potentially improving mood regulation and neurotransmitter function.


Sujet(s)
Alimentation riche en graisse , Hypercholestérolémie , Souris de lignée C57BL , Quinoléines , Transporteurs de la sérotonine , Animaux , Quinoléines/pharmacologie , Quinoléines/usage thérapeutique , Hypercholestérolémie/traitement médicamenteux , Hypercholestérolémie/métabolisme , Transporteurs de la sérotonine/métabolisme , Transporteurs de la sérotonine/génétique , Souris , Alimentation riche en graisse/effets indésirables , Mâle , Souris knockout , Récepteurs aux lipoprotéines LDL/métabolisme , Récepteurs aux lipoprotéines LDL/génétique , Tomographie par émission de positons , Cholestérol LDL/sang , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Encéphale/imagerie diagnostique , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/pharmacologie , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/usage thérapeutique
5.
Am J Reprod Immunol ; 92(2): e13918, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39158962

RÉSUMÉ

BACKGROUND: Zishen Qingre Lishi Huayu recipe (ZQLHR) has shown significant therapeutic effects in treating sex hormone levels and follicular developmental disorders in patients with polycystic ovary syndrome (PCOS). However, little is known about the potential mechanisms of its treatment. METHODS: Dehydroepiandrosterone and a high-fat diet induced the PCOS model rat. The serum of rats was collected to detect the levels of sex hormones and inflammatory cytokines by enzyme-linked immunosorbent assay, and the ovaries were collected for ovarian histopathology and qPCR assay to detect the levels of inflammatory cytokines in ovarian tissues. Granulosa cells (GCs) were collected for western blot assay to detect of IL-1ß, IL-6R, and LOX protein expression levels. RESULTS: ZQLHR could reduce body weight, regulate estrous cycles, and improve serum sex hormone levels, follicular development, and insulin resistance (IR) in PCOS model rats. In addition, ZQLHR treatment improved the levels of inflammatory cytokines in serum and ovary, and regulated the protein expression of IL-6R, IL-1ß, and LOX in GCs of PCOS model rats. The results showed that the HOMA-IR index increased with the increasing levels of IL-6, IL-1ß, and CRP, and decreased with the increased IL-10. CONCLUSION: This study reveals that the treatment of endocrine disorders and ovulation disorders in PCOS with ZQLHR may be closely related to the improvement of systemic and ovarian inflammation in PCOS patients, as well as the inhibition of IL-6R, IL-1ß, and LOX expression in GCs, which reemphasizes the role of reducing chronic inflammatory states in the treatment of PCOS. Moreover, this study reemphasizes the correlation between multiple inflammatory mediators and IR.


Sujet(s)
Modèles animaux de maladie humaine , Médicaments issus de plantes chinoises , Inflammation , Ovaire , Syndrome des ovaires polykystiques , Animaux , Femelle , Syndrome des ovaires polykystiques/traitement médicamenteux , Syndrome des ovaires polykystiques/métabolisme , Rats , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Inflammation/traitement médicamenteux , Ovaire/anatomopathologie , Ovaire/effets des médicaments et des substances chimiques , Ovaire/métabolisme , Rat Sprague-Dawley , Cytokines/métabolisme , Humains , Alimentation riche en graisse , Cellules de la granulosa/métabolisme , Cellules de la granulosa/effets des médicaments et des substances chimiques , Insulinorésistance , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/sang
6.
Sci Rep ; 14(1): 18724, 2024 08 12.
Article de Anglais | MEDLINE | ID: mdl-39134597

RÉSUMÉ

ATP6AP2 knockout in the renal nephron impairs receptor-mediated endocytosis, increasing urinary albumin and glucose excretion and impairing weight gain. Nonesterified fatty acids (NEFA) in urine are bound to albumin and reabsorbed in the proximal tubule through receptor-mediated endocytosis by the megalin-cubilin complex. We hypothesized that ATP6AP2 knockout increases urinary NEFA excretion through a reduction in megalin. Ten-week-old male C57BL/6 mice with nephron specific inducible ATP6AP2 knockout and noninduced controls were fed either normal diet (ND 12% fat) or high fat diet (HFD 45% fat) for 6 months. ATP6AP2 knockout significantly increased urine albumin:creatinine ratio in both ND and HFD fed mice while normalized urine NEFA concentration increased 489% and 259% in ND and HFD knockout mice compared to respective controls. Knockout decreased renal cortical megalin mRNA by 47% on ND and 49% on HFD while megalin protein expression decreased by 36% and 44% respectively. At the same time, markers of mTOR activity were increased while autophagy was impaired. Our results indicate that nephron specific ATP6AP2 knockout increases urinary NEFA excretion in the setting of impaired receptor-mediated endocytosis. Further investigation should determine whether ATP6AP2 contributes to obesity related ectopic lipid deposition in the proximal tubule.


Sujet(s)
Protéine-2 apparentée au récepteur des LDL , Néphrons , Animaux , Mâle , Souris , Alimentation riche en graisse , Acides gras/métabolisme , Acide gras libre/métabolisme , Cortex rénal/métabolisme , Protéine-2 apparentée au récepteur des LDL/génétique , Protéine-2 apparentée au récepteur des LDL/métabolisme , Souris de lignée C57BL , Souris knockout , Néphrons/métabolisme , Récepteur de la prorénine , Vacuolar Proton-Translocating ATPases/génétique , Vacuolar Proton-Translocating ATPases/métabolisme
7.
Clin Sci (Lond) ; 138(17): 1039-1054, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39136693

RÉSUMÉ

Maternal high-fat diet intake has profound effects on the long-term health of offspring, predisposing them to a higher susceptibility to obesity and metabolic dysfunction-associated steatotic liver disease. However, the detailed mechanisms underlying the role of a maternal high-fat diet in hepatic lipid accumulation in offspring, especially at the weaning age, remain largely unclear. In this study, female C57BL/6J mice were randomly assigned to either a high-fat diet or a control diet, and lipid metabolism parameters were assessed in male offspring at weaning. Gut microbiota analysis and targeted metabolomics of short-chain fatty acids (SCFAs) in these offspring were further performed. Both in vivo and in vitro studies were conducted to explore the role of butyrate in hepatic cholesterol excretion in the liver and HepG2 cells. Our results showed that maternal high-fat feeding led to obesity and dyslipidemia, and exacerbated hepatic lipid accumulation in the livers of offspring at weaning. We observed significant decreases in the abundance of the Firmicutes phylum and the Allobaculum genus, known as producers of SCFAs, particularly butyrate, in the offspring of dams fed a high-fat diet. Additionally, maternal high-fat diet feeding markedly decreased serum butyrate levels and down-regulated ATP-binding cassette transporters G5 (ABCG5) in the liver, accompanied by decreased phosphorylated AMP-activated protein kinase (AMPK) and histone deacetylase 5 (HADC5) expressions. Subsequent in vitro studies revealed that butyrate could induce ABCG5 activation and alleviate lipid accumulation via the AMPK-pHDAC5 pathway in HepG2 cells. Moreover, knockdown of HDAC5 up-regulated ABCG5 expression and promoted cholesterol excretion in HepG2 cells. In conclusion, our study provides novel insights into how maternal high-fat diet feeding inhibits hepatic cholesterol excretion and down-regulates ABCG5 through the butyrate-AMPK-pHDAC5 pathway in offspring at weaning.


Sujet(s)
Membre-5 de la sous-famille G des transporteurs à cassette liant l'ATP , Butyrates , Cholestérol , Alimentation riche en graisse , Microbiome gastro-intestinal , Foie , Souris de lignée C57BL , Animaux , Alimentation riche en graisse/effets indésirables , Femelle , Butyrates/métabolisme , Humains , Foie/métabolisme , Cellules HepG2 , Membre-5 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Membre-5 de la sous-famille G des transporteurs à cassette liant l'ATP/génétique , Mâle , Cholestérol/métabolisme , Cholestérol/sang , Grossesse , Souris , Métabolisme lipidique , Effets différés de l'exposition prénatale à des facteurs de risque/métabolisme , Phénomènes physiologiques nutritionnels maternels , Obésité/métabolisme , Obésité/microbiologie , Dyslipidémies/métabolisme , Dyslipidémies/microbiologie , Dyslipidémies/étiologie , Lipoprotéines
8.
Nutr Diabetes ; 14(1): 65, 2024 08 16.
Article de Anglais | MEDLINE | ID: mdl-39152116

RÉSUMÉ

BACKGROUND: Diet and gut microbiota contribute to non-alcoholic steatohepatitis (NASH) progression. High-fat diets (HFDs) change gut microbiota compositions, induce gut dysbiosis, and intestinal barrier leakage, which facilitates portal influx of pathogen-associated molecular patterns including lipopolysaccharides (LPS) to the liver and triggers inflammation in NASH. Current therapeutic drugs for NASH have adverse side effects; however, several foods and herbs that exhibit hepatoprotection could be an alternative method to prevent NASH. METHODS: We investigated ginger essential oil (GEO) against palm oil-containing HFDs in LPS-injected murine NASH model. RESULTS: GEO reduced plasma alanine aminotransferase levels and hepatic pro-inflammatory cytokine levels; and increased antioxidant catalase, glutathione reductase, and glutathione levels to prevent NASH. GEO alleviated hepatic inflammation through mediated NLR family pyrin domain-containing 3 (NLRP3) inflammasome and LPS/Toll-like receptor four (TLR4) signaling pathways. GEO further increased beneficial bacterial abundance and reduced NASH-associated bacterial abundance. CONCLUSION: This study demonstrated that GEO prevents NASH progression which is probably associated with the alterations of gut microbiota and inhibition of the LPS/TLR4/NF-κB pathway. Hence, GEO may offer a promising application as a dietary supplement for the prevention of NASH.


Sujet(s)
Microbiome gastro-intestinal , Inflammasomes , Lipopolysaccharides , Souris de lignée C57BL , Protéine-3 de la famille des NLR contenant un domaine pyrine , Stéatose hépatique non alcoolique , Huile essentielle , Transduction du signal , Récepteur de type Toll-4 , Zingiber officinale , Animaux , Stéatose hépatique non alcoolique/prévention et contrôle , Stéatose hépatique non alcoolique/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Récepteur de type Toll-4/métabolisme , Huile essentielle/pharmacologie , Huile essentielle/usage thérapeutique , Souris , Inflammasomes/métabolisme , Inflammasomes/effets des médicaments et des substances chimiques , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Alimentation riche en graisse/effets indésirables , Évolution de la maladie , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine
9.
Sci Rep ; 14(1): 19227, 2024 08 20.
Article de Anglais | MEDLINE | ID: mdl-39164365

RÉSUMÉ

Maternal malnutrition has been associated with neurodevelopmental deficits and long-term implications on the offspring's health and behavior. Here, we investigated the effects of maternal low-protein diet (LPD) or obesity-inducing maternal high-fat diet (HFD) on dyadic social interactions, group organization and autism-related behaviors in mice. We found that maternal HFD induced an autism-related behavioral phenotype in the male offspring, including a robust decrease in sociability, increased aggression, cognitive rigidity and repetitive behaviors. Maternal LPD led to a milder yet significant effect on autism-related symptoms, with no effects on olfactory-mediated social behavior. Under naturalistic conditions in a group setting, this manifested in altered behavioral repertoires, increased magnitude in dominance relations, and reduced interactions with novel social stimuli in the HFD male offspring, but not in the LPD offspring. Finally, we found HFD-induced transcriptomic changes in the olfactory bulbs of the male offspring. Together, our findings show that maternal malnutrition induces long-lasting effects on aggression and autism-related behaviors in male offspring, and potential impairments in brain regions processing chemosensory signals.


Sujet(s)
Trouble autistique , Comportement animal , Alimentation riche en graisse , Régime pauvre en protéines , Comportement social , Animaux , Alimentation riche en graisse/effets indésirables , Femelle , Mâle , Souris , Trouble autistique/étiologie , Trouble autistique/métabolisme , Grossesse , Régime pauvre en protéines/effets indésirables , Agressivité , Effets différés de l'exposition prénatale à des facteurs de risque/métabolisme , Souris de lignée C57BL , Phénomènes physiologiques nutritionnels maternels , Bulbe olfactif/métabolisme , Modèles animaux de maladie humaine , Obésité/métabolisme , Obésité/étiologie
10.
Reprod Biol Endocrinol ; 22(1): 105, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39164729

RÉSUMÉ

BACKGROUND: Obesity is a global health issue with detrimental effects on various human organs, including the reproductive system. Observational human data and several lines of animal experimental data suggest that maternal obesity impairs ovarian function and early embryo development, but the precise pathogenesis remains unclear. METHODS: We established a high-fat diet (HFD)-induced obese female mouse model to assess systemic metabolism, ovarian morphology, and oocyte function in mice. For the first time, this study employed single-cell RNA sequencing to explore the altered transcriptomic landscape of preimplantation embryos at different stages in HFD-induced obese mice. Differential gene expression analysis, enrichment analysis and protein-protein interactions network analysis were performed. RESULTS: HFD-induced obese female mice exhibited impaired glucolipid metabolism and insulin resistance. The ovaries of HFD mice had a reduced total follicle number, an increased proportion of atretic follicles, and irregular granulosa cell arrangement. Furthermore, the maturation rate of embryonic development by in vitro fertilization of oocytes was significantly decreased in HFD mice. Additionally, the transcriptional landscapes of preimplantation embryos at different stages in mice induced by different diets were significantly distinguished. The maternal-to-zygotic transition was also affected by the failure to remove maternal RNAs and to turn off zygotic genome expression. CONCLUSIONS: HFD-induced obesity impaired ovarian morphology and oocyte function in female mice and further led to alterations in the transcriptional landscape of preimplantation embryos at different stages of HFD mice.


Sujet(s)
Alimentation riche en graisse , Développement embryonnaire , Obésité , Ovocytes , Analyse de séquence d'ARN , Analyse sur cellule unique , Animaux , Femelle , Alimentation riche en graisse/effets indésirables , Ovocytes/métabolisme , Souris , Développement embryonnaire/génétique , Développement embryonnaire/effets des médicaments et des substances chimiques , Obésité/génétique , Obésité/métabolisme , Souris de lignée C57BL , Grossesse , Blastocyste/métabolisme
11.
Front Endocrinol (Lausanne) ; 15: 1440070, 2024.
Article de Anglais | MEDLINE | ID: mdl-39145314

RÉSUMÉ

Previous studies indicate that CNS administration of oxytocin (OT) reduces body weight in high fat diet-induced obese (DIO) rodents by reducing food intake and increasing energy expenditure (EE). We recently demonstrated that hindbrain (fourth ventricular [4V]) administration of OT elicits weight loss and elevates interscapular brown adipose tissue temperature (TIBAT, a surrogate measure of increased EE) in DIO mice. What remains unclear is whether OT-elicited weight loss requires increased sympathetic nervous system (SNS) outflow to IBAT. We hypothesized that OT-induced stimulation of SNS outflow to IBAT contributes to its ability to activate BAT and elicit weight loss in DIO mice. To test this hypothesis, we determined the effect of disrupting SNS activation of IBAT on the ability of 4V OT administration to increase TIBAT and elicit weight loss in DIO mice. We first determined whether bilateral surgical SNS denervation to IBAT was successful as noted by ≥ 60% reduction in IBAT norepinephrine (NE) content in DIO mice. NE content was selectively reduced in IBAT at 1-, 6- and 7-weeks post-denervation by 95.9 ± 2.0, 77.4 ± 12.7 and 93.6 ± 4.6% (P<0.05), respectively and was unchanged in inguinal white adipose tissue, pancreas or liver. We subsequently measured the effects of acute 4V OT (1, 5 µg ≈ 0.99, 4.96 nmol) on TIBAT in DIO mice following sham or bilateral surgical SNS denervation to IBAT. We found that the high dose of 4V OT (5 µg ≈ 4.96 nmol) elevated TIBAT similarly in sham mice as in denervated mice. We subsequently measured the effects of chronic 4V OT (16 nmol/day over 29 days) or vehicle infusions on body weight, adiposity and food intake in DIO mice following sham or bilateral surgical denervation of IBAT. Chronic 4V OT reduced body weight by 5.7 ± 2.23% and 6.6 ± 1.4% in sham and denervated mice (P<0.05), respectively, and this effect was similar between groups (P=NS). OT produced corresponding reductions in whole body fat mass (P<0.05). Together, these findings support the hypothesis that sympathetic innervation of IBAT is not necessary for OT-elicited increases in BAT thermogenesis and reductions of body weight and adiposity in male DIO mice.


Sujet(s)
Tissu adipeux brun , Adiposité , Alimentation riche en graisse , Souris de lignée C57BL , Obésité , Ocytocine , Système nerveux sympathique , Animaux , Ocytocine/pharmacologie , Tissu adipeux brun/effets des médicaments et des substances chimiques , Tissu adipeux brun/métabolisme , Tissu adipeux brun/innervation , Mâle , Souris , Obésité/métabolisme , Système nerveux sympathique/effets des médicaments et des substances chimiques , Alimentation riche en graisse/effets indésirables , Adiposité/effets des médicaments et des substances chimiques , Poids/effets des médicaments et des substances chimiques , Perte de poids/effets des médicaments et des substances chimiques , Souris obèse , Métabolisme énergétique/effets des médicaments et des substances chimiques , Norépinéphrine/métabolisme
12.
Gut Microbes ; 16(1): 2390164, 2024.
Article de Anglais | MEDLINE | ID: mdl-39154362

RÉSUMÉ

Non-alcoholic fatty liver disease (NAFLD) has emerged as a global health concern, lacking specific therapeutic strategies. Time-restricted feeding (TRF) regimen demonstrated beneficial effects in NAFLD; however, the underlying mechanisms remain unclear. In this study, we established a NAFLD mouse model through a high-fat diet (HFD) and implemented the 16:8 TRF regimen for a duration of 6 weeks. We demonstrated that TRF remarkably alleviated hepatic steatosis in HFD mice. Of note, aldehyde oxidase 1 (AOX1), a key enzyme in hepatic nicotinamide (NAM) catabolism, exhibited apparent upregulation in response to HFD, leading to abnormal accumulation of N-Methyl-6-pyridone-3-carboxamide (N-Me-6-PY, also known as 2PY) and N-Methyl-4-pyridone-5-carboxamide (N-Me-4-PY, also known as 4PY), whereas it was almost restored by TRF. Both N-Me-6-PY and N-Me-4-PY promoted de novo lipogenesis and fatty acid uptake capacities in hepatocyte, and aggravated hepatic steatosis in mice either fed chow diet or HFD. In contrast, pharmacological inhibition of AOX1 was sufficient to ameliorate the hepatic steatosis and lipid metabolic dysregulation induced by HFD. Moreover, transplantation of fecal microbiota efficiently mimicked the modulatory effect of TRF on NAM metabolism, thus mitigating hepatic steatosis and lipid metabolic disturbance, suggesting a gut microbiota-dependent manner. In conclusion, our study reveals the intricate relationship between host NAM metabolic modification and gut microbiota remodeling during the amelioration of NAFLD by TRF, providing promising insights into the prevention and treatment of NAFLD.


Sujet(s)
Alimentation riche en graisse , Microbiome gastro-intestinal , Foie , Souris de lignée C57BL , Nicotinamide , Stéatose hépatique non alcoolique , Animaux , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/microbiologie , Souris , Foie/métabolisme , Alimentation riche en graisse/effets indésirables , Mâle , Nicotinamide/métabolisme , Modèles animaux de maladie humaine , Métabolisme lipidique , Aldehyde oxidase/métabolisme , Lipogenèse , Hépatocytes/métabolisme , Humains
13.
Sci Rep ; 14(1): 18602, 2024 08 10.
Article de Anglais | MEDLINE | ID: mdl-39127712

RÉSUMÉ

Consumption of high-caloric diets contributes to the alarming number of overweight and obese individuals worldwide, which in turn leads to several diseases and multiple organ dysfunction. Not only has the number of calories taken per day but also the type of fat in the diet has an important impact on health. Accordingly, the purpose of the current study was to examine the impact of different types of high-caloric fat diets on the metabolic status and the integrity of the liver and aorta in albino rats. Adult male albino rats were divided into 6 groups: Control group, long chain-saturated fat group (SFD), long chain-monounsaturated fat (MUFAs) group, long chain-polyunsaturated fat (PUFAs) group, medium-chain fat (MCFAs) group, and short-chain fat (SCFAs) group. Body mass index (BMI), Lee index, and visceral fat amount were reported. Serum levels of insulin, liver transaminases, lipid profile, and different oxidative stress and inflammatory markers were evaluated. Homeostasis Model Assessment of Insulin Resistance (HOMA-IR), and adiponectin/leptin ratio were also calculated. Histopathological examinations of liver and aorta with Masson's trichrome stain, and immune-staining for Nuclear Factor Erythroid-2-Related Factor-2 (Nrf2) were also done. SFD group showed significantly elevated liver transaminases, inflammatory markers, HOMA-IR, dyslipidemia, reduced adiponectin, and deficient anti-oxidative response compared to other groups together with disturbed hepatic and aortic architecture. Other treated groups showed an improvement. PUFAs group showed the highest level of improvement. Not all high-fat diets are hazardous. Diets rich in PUFAs, MUFAs, MCFAs, or SCFAs may protect against the hazards of high caloric diet.


Sujet(s)
Aorte , Alimentation riche en graisse , Foie , Animaux , Foie/métabolisme , Foie/anatomopathologie , Rats , Mâle , Alimentation riche en graisse/effets indésirables , Aorte/métabolisme , Aorte/anatomopathologie , Stress oxydatif , Insulinorésistance , Insuline/sang , Insuline/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme
14.
Sci Rep ; 14(1): 18641, 2024 08 11.
Article de Anglais | MEDLINE | ID: mdl-39128931

RÉSUMÉ

There are genetic and environmental risk factors that contribute to the development of cognitive decline in Alzheimer's disease (AD). Some of these include the genetic predisposition of the apolipoprotein E4 genotype, consuming a high-fat diet (HFD), and the female sex. Brain insulin receptor resistance and deficiency have also been shown to be associated with AD and cognitive impairment. Intranasal (INL) insulin enhances cognition in AD, but the response varies due to genotype, diet, and sex. We investigated here the combination of these risk factors in a humanized mouse model, expressing E3 or E4, following a HFD in males and females on cognitive performance and the brain distribution of insulin following INL delivery. The HFD had a negative effect on survival in male mice only, requiring sex to be collapsed. We found many genotype, diet, and genotype x diet effects in anxiety-related tasks. We further found beneficial effects of INL insulin in our memory tests, with the most important findings showing a beneficial effect of INL insulin in mice on a HFD. We found insulin distribution throughout the brain after INL delivery was largely unaffected by diet and genotype, indicating these susceptible groups can still receive adequate levels of insulin following INL delivery. Our findings support the involvement of brain insulin signaling in cognition and highlight continuing efforts investigating mechanisms resulting from treatment with INL insulin.


Sujet(s)
Administration par voie nasale , Encéphale , Cognition , Alimentation riche en graisse , Insuline , Animaux , Femelle , Mâle , Insuline/métabolisme , Alimentation riche en graisse/effets indésirables , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Cognition/effets des médicaments et des substances chimiques , Souris , Humains , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme , Souris transgéniques , Maladie d'Alzheimer/métabolisme , Modèles animaux de maladie humaine
15.
Sci Rep ; 14(1): 18377, 2024 08 08.
Article de Anglais | MEDLINE | ID: mdl-39112593

RÉSUMÉ

The effects of calcitonin gene-related peptide (CGRP) on atherosclerosis remain unclear. We used apolipoprotein E-deficient (ApoE-/-) mice to generate double-knockout ApoE-/-:CGRP-/- mice lacking alpha CGRP. ApoE-/-:CGRP-/- mice exhibited larger atherosclerotic plaque areas, peritoneal macrophages with enhanced migration functions, and elevated levels of the inflammatory cytokine tumor necrosis factor (TNF)-⍺. Thus, we also explored whether inhibiting TNF-⍺ could improve atherosclerosis in ApoE-/-:CGRP-/- mice by administering etanercept intraperitoneally once a week (5 mg/kg) alongside a high-fat diet for 2 weeks. This treatment led to significant reductions in aortic root lesion size, atherosclerotic plaque area and macrophage migration in ApoE-/-:CGRP-/- mice compared with mice treated with human IgG (5 mg/kg). We further examined whether results observed in ApoE-/-:CGRP-/- mice could similarly be obtained by administering a humanized monoclonal CGRP antibody, galcanezumab, to ApoE-/- mice. ApoE-/- mice were subcutaneously administered galcanezumab at an initial dose of 50 mg/kg, followed by a dose of 30 mg/kg in the second week. Galcanezumab administration did not affect systolic blood pressure, serum lipid levels, or macrophage migration but led to a significant increase in lipid deposition at the aortic root. These findings suggest that alpha CGRP plays a critical role in inhibiting the progression of atherosclerosis.


Sujet(s)
Apolipoprotéines E , Athérosclérose , Peptide relié au gène de la calcitonine , Souris knockout , Plaque d'athérosclérose , Animaux , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Peptide relié au gène de la calcitonine/métabolisme , Souris , Apolipoprotéines E/déficit , Apolipoprotéines E/génétique , Plaque d'athérosclérose/anatomopathologie , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/génétique , Alimentation riche en graisse/effets indésirables , Facteur de nécrose tumorale alpha/métabolisme , Mâle , Souris invalidées pour les gènes ApoE , Modèles animaux de maladie humaine , Humains , Anticorps monoclonaux humanisés/pharmacologie , Étanercept/pharmacologie , Souris de lignée C57BL , Mouvement cellulaire/effets des médicaments et des substances chimiques , Aorte/métabolisme , Aorte/anatomopathologie , Aorte/effets des médicaments et des substances chimiques
16.
J Cell Biol ; 223(10)2024 Oct 07.
Article de Anglais | MEDLINE | ID: mdl-39150520

RÉSUMÉ

The integrated stress response (ISR) is a vital signaling pathway initiated by four kinases, PERK, GCN2, HRI and PKR, that ensure cellular resilience and protect cells from challenges. Here, we investigated whether increasing ISR signaling could rescue diabetes-like phenotypes in a mouse model of diet-induced obesity (DIO). We show that the orally available and clinically approved GCN2 activator halofuginone (HF) can activate the ISR in mouse tissues. We found that daily oral administration of HF increases glucose tolerance whilst reducing weight gain, insulin resistance, and serum insulin in DIO mice. Conversely, the ISR inhibitor GSK2656157, used at low doses to optimize its selectivity, aggravates glucose intolerance in DIO mice. Whilst loss of function mutations in mice and humans have revealed that PERK is the essential ISR kinase that protects from diabetes, our work demonstrates the therapeutic value of increasing ISR signaling by activating the related kinase GCN2 to reduce diabetes phenotypes in a DIO mouse model.


Sujet(s)
Obésité , Phénotype , Pipéridines , Protein-Serine-Threonine Kinases , Quinazolinones , Transduction du signal , eIF-2 Kinase , Animaux , Quinazolinones/pharmacologie , Pipéridines/pharmacologie , Souris , eIF-2 Kinase/métabolisme , eIF-2 Kinase/génétique , Obésité/anatomopathologie , Obésité/métabolisme , Obésité/prévention et contrôle , Obésité/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Souris de lignée C57BL , Mâle , Insulinorésistance , Insuline/métabolisme , Insuline/sang , Stress physiologique/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Alimentation riche en graisse/effets indésirables , Diabète/anatomopathologie , Diabète/métabolisme , Diabète/génétique , Diabète/traitement médicamenteux , Diabète/prévention et contrôle , Intolérance au glucose/traitement médicamenteux , Adénine/analogues et dérivés , Indoles
17.
PeerJ ; 12: e17891, 2024.
Article de Anglais | MEDLINE | ID: mdl-39148685

RÉSUMÉ

Objective: This study aims to investigate the effects of semaglutide on gut microbiota, cognitive function, and inflammation in obese mice. Method: Twenty-four C57BL/6J male mice were randomly assigned to three groups: a normal-chow diet group (NCD, n = 8), high-fat diet group (HFD, n = 8), and HFD+semaglutide group (Sema, n = 8). The mice were fed a HFD to establish an animal model of obesity and then administered with semaglutide or saline for 12 weeks. Cognitive function was assessed using the Morris water maze test. Serum pro-inflammatory cytokines were measured. 16S rRNA gene sequencing technology was used to explore gut microbiota characteristics in obese mice. Result: Obese mice showed significant cognitive impairment and inflammation. Semaglutide improved cognitive function and attenuated inflammation induced by a HFD diet. The abundance of gut microbiota was significantly changed in the HFD group, including decreased Akkermansia, Muribaculaceae, Coriobacteriaceae_UCG_002, Clostridia_UCG_014 and increased Romboutsia, Dubosiella, Enterorhabdus. Whereas semaglutide could dramatically reverse the relative abundance of these gut microbiota. Correlation analysis suggested that cognitive function was positively correlated with Muribaculaceae and Clostridia_UCG_014, and negatively associated with Romboutsia and Dubosiella. Romboutsia was positively correlated with TNFα, IL-6 and IL-1ß. While Clostridia_UCG_014 was negatively related to TNFα, IL-6 and IL-1ß. Conclusions: For the first time semaglutide displayed different regulatory effects on HFD-induced gut microbiota dysbiosis. Semaglutide could regulate the structure and composition of gut microbiota associated with cognitive function and inflammation. Thus, affecting gut microbiota might be a potential mechanism of semaglutide in attenuating cognitive function and inflammation.


Sujet(s)
Cognition , Alimentation riche en graisse , Microbiome gastro-intestinal , Peptides glucagon-like , Inflammation , Souris de lignée C57BL , Obésité , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Peptides glucagon-like/pharmacologie , Souris , Mâle , Obésité/microbiologie , Obésité/traitement médicamenteux , Inflammation/traitement médicamenteux , Cognition/effets des médicaments et des substances chimiques , Alimentation riche en graisse/effets indésirables , Souris obèse , Modèles animaux de maladie humaine , Cytokines/métabolisme , Cytokines/sang , Dysfonctionnement cognitif/traitement médicamenteux
18.
Cell Mol Life Sci ; 81(1): 329, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090270

RÉSUMÉ

Decidualisation of the endometrium is a key event in early pregnancy, which enables embryo implantation. Importantly, the molecular processes impairing decidualisation in obese mothers are yet to be characterised. We hypothesise that impaired decidualisation in obese mice is mediated by the upregulation of leptin modulators, the suppressor of cytokine signalling 3 (SOCS3) and the protein tyrosine phosphatase non-receptor type 2 (PTPN2), together with the disruption of progesterone (P4)-signal transducer and activator of transcription (STAT3) signalling. After feeding mice with chow diet (CD) or high-fat diet (HFD) for 16 weeks, we confirmed the downregulation of P4 and oestradiol (E2) steroid receptors in decidua from embryonic day (E) 6.5 and decreased proliferation of stromal cells from HFD. In vitro decidualised mouse endometrial stromal cells (MESCs) and E6.5 deciduas from the HFD showed decreased expression of decidualisation markers, followed by the upregulation of SOCS3 and PTPN2 and decreased phosphorylation of STAT3. In vivo and in vitro leptin treatment of mice and MESCs mimicked the results observed in the obese model. The downregulation of Socs3 and Ptpn2 after siRNA transfection of MESCs from HFD mice restored the expression level of decidualisation markers. Finally, DIO mice placentas from E18.5 showed decreased labyrinth development and vascularisation and fetal growth restricted embryos. The present study revealed major defects in decidualisation in obese mice, characterised by altered uterine response to E2 and P4 steroid signalling. Importantly, altered hormonal response was associated with increased expression of leptin signalling modulators SOCS3 and PTPN2. Elevated levels of SOCS3 and PTPN2 were shown to molecularly affect decidualisation in obese mice, potentially disrupting the STAT3-PR regulatory molecular hub.


Sujet(s)
Caduques , Retard de croissance intra-utérin , Leptine , Placenta , Transduction du signal , Animaux , Femelle , Souris , Grossesse , Caduques/métabolisme , Caduques/anatomopathologie , Alimentation riche en graisse/effets indésirables , Retard de croissance intra-utérin/métabolisme , Retard de croissance intra-utérin/anatomopathologie , Leptine/métabolisme , Souris de lignée C57BL , Souris obèse , Obésité/métabolisme , Obésité/anatomopathologie , Placenta/métabolisme , Progestérone/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 2/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 2/génétique , Facteur de transcription STAT-3/métabolisme , Cellules stromales/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique
19.
Chem Commun (Camb) ; 60(68): 9089-9092, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39108142

RÉSUMÉ

Herein, we have developed a non-enzymatic, isothermal amplification assay (NIA sensor) based on a catalytic hairpin assembly (CHA) reaction for quantifying the relative abundance of Akkermansia muciniphila. Through detection of the MUC-1437 gene (limit of detection: 8.3 fM) in a dynamic range from 10 fM to 1 nM, the NIA sensor shows high sensitivity and selectivity in preclinical models of mice fed a normal or high-fat diet (HFD), and treated with antibiotics (ATB). The NIA sensor, which operates without the use of any enzymes, leading to simplicity and cost-effectiveness, has great potential for biosensing research and clinical diagnostic applications.


Sujet(s)
Akkermansia (genre) , Techniques d'amplification d'acides nucléiques , Animaux , Souris , Techniques de biocapteur , Antibactériens/pharmacologie , Alimentation riche en graisse , Limite de détection , Verrucomicrobia/génétique , Verrucomicrobia/isolement et purification
20.
Gene ; 929: 148812, 2024 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-39116959

RÉSUMÉ

Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease and represents the main cause of liver cirrhosis and hepatocellular carcinoma. Cav3.2 is a T-type calcium channel that is widely present in tissues throughout the body and plays a vital role in energy and metabolic balance. However, the effects of Cav3.2 on the NFALD remain unclear. Here, we investigated the role of Cav3.2 channel in the development and progression of NAFLD. After 16 weeks on a high-fat diets (HFD), Cav3.2 knockout (Cav3.2 KO) improved hepatic steatosis, liver injury and metabolic syndrome in an NAFLD mouse model. We provided evidence that Cav3.2 KO inhibited HFD-induced hepatic oxidative stress, inflammation and hepatocyte apoptosis. In addition, Cav3.2 KO also attenuated hepatic lipid accumulation, oxidative stress, inflammation and hepatocyte apoptosis in palmitic acid/oleic acid (PAOA)-treated primary hepatocytes. These results suggest that therapeutic approaches targeting Cav3.2 provide effective approaches for treating NAFLD.


Sujet(s)
Apoptose , Canaux calciques de type T , Alimentation riche en graisse , Hépatocytes , Souris knockout , Stéatose hépatique non alcoolique , Stress oxydatif , Animaux , Stéatose hépatique non alcoolique/génétique , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Canaux calciques de type T/génétique , Canaux calciques de type T/métabolisme , Souris , Alimentation riche en graisse/effets indésirables , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Mâle , Souris de lignée C57BL , Modèles animaux de maladie humaine , Foie/métabolisme , Foie/anatomopathologie , Inflammation/génétique , Inflammation/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE