Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.497
Filtrer
1.
Cell Rep ; 43(7): 114425, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38970789

RÉSUMÉ

Obesity is a global health challenge with limited therapeutic solutions. Here, we demonstrate the engineering of an energy-dissipating hybrid tissue (EDHT) in the body for weight control. EDHT is constructed by implanting a synthetic gel matrix comprising immunomodulatory signals and functional cells into the recipient mouse. The immunomodulatory signals induce the host stromal cells to create an immunosuppressive niche that protects the functional cells, which are overexpressing the uncoupling protein 1 (UCP1), from immune rejection. Consequently, these endogenous and exogenous cells co-develop a hybrid tissue that sustainedly produces UCP1 to accelerate the host's energy expenditure. Systematic experiments in high-fat diet (HFD) and transgenic (ob/ob) mice show that EDHT efficiently reduces body weight and relieves obesity-associated pathological conditions. Importantly, an 18-month observation for safety assessment excludes cell leakage from EDHT and reports no adverse physiological responses. Overall, EDHT demonstrates convincing efficacy and safety in controlling body weight.


Sujet(s)
Alimentation riche en graisse , Métabolisme énergétique , Obésité , Animaux , Obésité/métabolisme , Obésité/thérapie , Souris , Protéine-1 de découplage/métabolisme , Ingénierie tissulaire/méthodes , Mâle , Souris de lignée C57BL , Souris transgéniques , Humains , Poids , Souris obèse
2.
J Med Chem ; 67(14): 12439-12458, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38996004

RÉSUMÉ

The discovery of effective and safe antiobesity agents remains a challenging yet promising field. Our previous studies identified Bouchardatine derivatives as potential antiobesity agents. However, the 8a-aldehyde moiety rendered them unsuitable for drug development. In this study, we designed two series of novel derivatives to modify this structural feature. Through a structure-activity relationship study, we elucidated the role of the 8a-aldehyde group in toxicity induction. We identified compound 14d, featuring an 8a-N-acylhydrazone moiety, which exhibited significant lipid-lowering activity and reduced toxicity. Compound 14d shares a similar lipid-lowering mechanism with our lead compound 3, but demonstrates improved pharmacokinetic properties and safety profile. Both oral and injectable administration of 14d significantly reduced body weight gain and ameliorated metabolic syndrome in diet-induced obese mice. Our findings identify 14d as a promising antiobesity agent and highlight the potential of substituting the aldehyde group with an N-acylhydrazone to enhance drug-like properties.


Sujet(s)
Aldéhydes , Agents antiobésité , Hydrazones , Obésité , Animaux , Agents antiobésité/pharmacologie , Agents antiobésité/synthèse chimique , Agents antiobésité/pharmacocinétique , Agents antiobésité/usage thérapeutique , Agents antiobésité/composition chimique , Hydrazones/pharmacologie , Hydrazones/composition chimique , Hydrazones/synthèse chimique , Hydrazones/pharmacocinétique , Hydrazones/usage thérapeutique , Souris , Relation structure-activité , Aldéhydes/composition chimique , Mâle , Obésité/traitement médicamenteux , Souris de lignée C57BL , Alimentation riche en graisse/effets indésirables , Humains , Souris obèse , Structure moléculaire
3.
J Med Chem ; 67(14): 11769-11788, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39013015

RÉSUMÉ

Here, we describe the development of the FGF21 analog zalfermin (NNC0194-0499, 15), intended for once-weekly sc dosing. Protein engineering was needed to address inherent druggability issues of the natural FGF21 hormone. Thus, deamidation of Asp121 was solved by mutation to glutamine, and oxidation of Met168 was solved by mutation to leucine. N-terminal region degradation by dipeptidyl peptidase IV was prevented by alanine residue elongation. To prevent inactivating metabolism by fibroblast activation protein and carboxypeptidase-like activity in the C-terminal region, and to achieve t1/2 extension (53 h in cynomolgus monkeys), we introduced a C18 fatty diacid at the penultimate position 180. The fatty diacid binds albumin in a reversible manner, such that the free fraction of zalfermin potently activates the FGF-receptor complex and retains receptor selectivity compared with FGF21, providing strong efficacy on body weight loss in diet-induced obese mice. Zalfermin is currently being clinically evaluated for the treatment of metabolic dysfunction-associated steatohepatitis.


Sujet(s)
Facteurs de croissance fibroblastique , Macaca fascicularis , Facteurs de croissance fibroblastique/métabolisme , Animaux , Souris , Humains , Mâle , Protéolyse/effets des médicaments et des substances chimiques , Souris obèse , Souris de lignée C57BL , Obésité/traitement médicamenteux , Obésité/métabolisme
4.
Obesity (Silver Spring) ; 32(8): 1483-1493, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39045674

RÉSUMÉ

OBJECTIVE: Thyroid hormone influences key metabolic pathways, and reduced sensitivity to thyroid hormone is considered a new risk factor for adverse metabolic outcomes. However, the association between thyroid hormone resistance and obesity in euthyroid individuals is still unknown. METHODS: We enrolled 8021 euthyroid individuals, calculated thyroid hormone resistance indices, and analyzed the association between thyroid hormone resistance and obesity by regression analysis. Furthermore, we conducted the thyrotropin-releasing hormone stimulation test in both control and obese mice (n = 5) to demonstrate the association. RESULTS: The euthyroid adults with overweight and obesity had increased thyroid hormone resistance indices (all p < 0.05). BMI and prevalence of overweight and obesity increased (odds ratio of thyroid feedback quantile-based index [ORTFQI] = 1.164, p = 0.036; OR of free triiodothyronine/free thyroxine [ORFT3/FT4] = 1.508, p < 0.001) following the elevation of thyroid hormone resistance indices. Mediation analysis indicated a complete mediation effect (beta coefficient of indirect effect [ßInd]= 6.838, p < 0.001) of metabolic disorders in the relationship. Furthermore, in the mice with obesity, the thyrotropin response to thyrotropin-releasing hormone stimulation (68.33-90.89 pg/mL) was comparatively blunted (p = 0.029). CONCLUSIONS: Euthyroid individuals with obesity exhibit both central and peripheral thyroid hormone resistance, a phenomenon that is more pronounced in individuals with metabolic abnormalities. Thyroid hormone resistance is associated with an increased prevalence of overweight and obesity mediated by metabolic disorders.


Sujet(s)
Obésité , Animaux , Souris , Études transversales , Mâle , Humains , Femelle , Adulte , Adulte d'âge moyen , Indice de masse corporelle , Tri-iodothyronine/sang , Hormones thyroïdiennes/sang , Syndrome de résistance aux hormones thyroïdiennes/épidémiologie , Thyroxine/sang , Hormone de libération de la thyréostimuline , Souris obèse , Thyréostimuline/sang , Souris de lignée C57BL , Surpoids
5.
Cell Mol Life Sci ; 81(1): 309, 2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39060446

RÉSUMÉ

The circadian clock system coordinates metabolic, physiological, and behavioral functions across a 24-h cycle, crucial for adapting to environmental changes. Disruptions in circadian rhythms contribute to major metabolic pathologies like obesity and Type 2 diabetes. Understanding the regulatory mechanisms governing circadian control is vital for identifying therapeutic targets. It is well characterized that chromatin remodeling and 3D structure at genome regulatory elements contributes to circadian transcriptional cycles; yet the impact of rhythmic chromatin topology in metabolic disease is largely unexplored. In this study, we explore how the spatial configuration of the genome adapts to diet, rewiring circadian transcription and contributing to dysfunctional metabolism. We describe daily fluctuations in chromatin contacts between distal regulatory elements of metabolic control genes in livers from lean and obese mice and identify specific lipid-responsive regions recruiting the clock molecular machinery. Interestingly, under high-fat feeding, a distinct interactome for the clock-controlled gene Dbp strategically promotes the expression of distal metabolic genes including Fgf21. Alongside, new chromatin loops between regulatory elements from genes involved in lipid metabolism control contribute to their transcriptional activation. These enhancers are responsive to lipids through CEBPß, counteracting the circadian repressor REVERBa. Our findings highlight the intricate coupling of circadian gene expression to a dynamic nuclear environment under high-fat feeding, supporting a temporally regulated program of gene expression and transcriptional adaptation to diet.


Sujet(s)
Chromatine , Horloges circadiennes , Acides gras , Foie , Souris de lignée C57BL , Souris obèse , Obésité , Animaux , Chromatine/métabolisme , Chromatine/génétique , Foie/métabolisme , Souris , Horloges circadiennes/génétique , Obésité/métabolisme , Obésité/génétique , Acides gras/métabolisme , Mâle , Alimentation riche en graisse/effets indésirables , Assemblage et désassemblage de la chromatine , Rythme circadien/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Métabolisme lipidique/génétique , Facteurs de croissance fibroblastique/métabolisme , Facteurs de croissance fibroblastique/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme
6.
Biol Direct ; 19(1): 52, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956667

RÉSUMÉ

BACKGROUND: Adiposity profoundly impacts reproductive health in both humans and animals. However, the precise subpopulations contributing to infertility under obese conditions remain elusive. RESULTS: In this study, we established an obese mouse model through an eighteen-week high-fat diet regimen in adult female mice. Employing single-cell RNA sequencing (scRNA-seq), we constructed a comprehensive single-cell atlas of ovarian tissues from these mice to scrutinize the impact of obesity on the ovarian microenvironment. ScRNA-seq revealed notable alterations in the microenvironment of ovarian tissues in obese mice. Granulosa cells, stromal cells, T cells, and macrophages exhibited functional imbalances compared to the control group. We observed heightened interaction strength in the SPP1-CD44 pairing within lgfbp7+ granulosa cell subtypes and Il1bhigh monocyte subtypes in the ovarian tissues of obese mice. Moreover, the interaction strength between Il1bhigh monocyte subtypes and Pdgfrb+ stromal cell subtypes in the form of TNF - TNFrsf1α interaction was also enhanced subsequently to obesity, potentially contributing to ovarian fibrosis pathogenesis. CONCLUSIONS: We propose a model wherein granulosa cells secrete SPP1 to activate monocytes, subsequently triggering TNF-α secretion by monocytes, thereby activating stromal cells and ultimately leading to the development of ovarian fibrosis. Intervening in this process may represent a promising avenue for improving clinical outcomes in fertility treatments for obese women.


Sujet(s)
Fibrose , Souris obèse , Obésité , Analyse sur cellule unique , Animaux , Femelle , Souris , Fibrose/génétique , Obésité/génétique , Obésité/métabolisme , Analyse de profil d'expression de gènes , Ovaire/métabolisme , Transcriptome , Souris de lignée C57BL , Alimentation riche en graisse/effets indésirables , Cellules de la granulosa/métabolisme
7.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(3): 349-358, 2024 Mar 28.
Article de Anglais, Chinois | MEDLINE | ID: mdl-38970508

RÉSUMÉ

OBJECTIVES: Obesity related glomerulopathy (ORG) is induced by obesity, but the pathogenesis remains unclear. This study aims to investigate the expression of early growth response protein 3 (EGR3) in the renal cortex tissues of ORG patients and high-fat diet-induced obese mice, and to further explore the molecular mechanism of EGR3 in inhibiting palmitic acid (PA) induced human podocyte inflammatory damage. METHODS: Renal cortex tissues were collected from ORG patients (n=6) who have been excluded from kidney damage caused by other diseases and confirmed by histopathology, and from obese mice induced by high-fat diet (n=10). Human and mouse podocytes were intervened with 150 µmol/L PA for 48 hours. EGR3 was overexpressed or silenced in human podocytes. Enzyme linked immunosorbent assay (ELISA) was used to detcet the levels of interleukin-6 (IL-6) and interleukin-1ß (IL-1ß). Real-time RT-PCR was used to detect the mRNA expressions of EGR3, podocytes molecular markers nephrosis 1 (NPHS1), nephrosis 2 (NPHS2), podocalyxin (PODXL), and podoplanin (PDPN). RNA-seq was performed to detect differentially expressed genes (DEGs) after human podocytes overexpressing EGR3 and treated with 150 µmol/L PA compared with the control group. Co-immunoprecipitation (Co-IP) combined with liquid chromatography tandem mass spectrometry (LC-MS) was used to detect potential interacting proteins of EGR3 and the intersected with the RNA-seq results. Co-IP confirmed the interaction between EGR3 and protein arginine methyltransferases 1 (PRMT1), after silencing EGR3 and PRMT1 inhibitor intervention, the secretion of IL-6 and IL-1ß in PA-induced podocytes was detected. Western blotting was used to detect the expression of phosphorylated signal transducer and activator of transcription 3 (p-STAT3) after overexpression or silencing of EGR3. RESULTS: EGR3 was significantly upregulated in renal cortex tissues of ORG patients and high-fat diet-induced obese mice (both P<0.01). In addition, after treating with 150 µmol/L PA for 48 hours, the expression of EGR3 in human and mouse podocytes was significantly upregulated (both P<0.05). Overexpression or silencing of EGR3 in human podocytes inhibited or promoted the secretion of IL-6 and IL-1ß in the cell culture supernatant after PA intervention, respectively, and upregulated or downregulated the expression of NPHS1, PODXL, NPHS2,and PDPN (all P<0.05). RNA-seq showed a total of 988 DEGs, and Co-IP+LC-MS identified a total of 238 proteins that may interact with EGR3. Co-IP confirmed that PRMT1 was an interacting protein with EGR3. Furthermore, PRMT1 inhibitors could partially reduce PA-induced IL-6 and IL-1ß secretion after EGR3 silencing in human podocytes (both P<0.05). Overexpression or silencing of EGR3 negatively regulated the expression of PRMT1 and p-STAT3. CONCLUSIONS: EGR3 may reduce ORG podocyte inflammatory damage by inhibiting the PRMT1/p-STAT3 pathway.


Sujet(s)
Facteur de transcription EGR-3 , Obésité , Podocytes , Protein-arginine N-methyltransferases , Protéines de répression , Facteur de transcription STAT-3 , Podocytes/métabolisme , Podocytes/anatomopathologie , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Animaux , Humains , Souris , Facteur de transcription STAT-3/métabolisme , Obésité/complications , Obésité/métabolisme , Facteur de transcription EGR-3/métabolisme , Facteur de transcription EGR-3/génétique , Protéines de répression/métabolisme , Protéines de répression/génétique , Transduction du signal , Maladies du rein/métabolisme , Maladies du rein/étiologie , Maladies du rein/anatomopathologie , Acide palmitique/pharmacologie , Alimentation riche en graisse/effets indésirables , Inflammation/métabolisme , Souris obèse , Mâle , Interleukine-1 bêta/métabolisme , Souris de lignée C57BL , Interleukine-6/métabolisme , Interleukine-6/génétique , Cortex rénal/métabolisme , Cortex rénal/anatomopathologie
8.
Proc Natl Acad Sci U S A ; 121(28): e2318691121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38968121

RÉSUMÉ

Dietary lipids play an essential role in regulating the function of the gut microbiota and gastrointestinal tract, and these luminal interactions contribute to mediating host metabolism. Palmitic Acid Hydroxy Stearic Acids (PAHSAs) are a family of lipids with antidiabetic and anti-inflammatory properties, but whether the gut microbiota contributes to their beneficial effects on host metabolism is unknown. Here, we report that treating chow-fed female and male germ-free (GF) mice with PAHSAs improves glucose tolerance, but these effects are lost upon high fat diet (HFD) feeding. However, transfer of feces from PAHSA-treated, but not vehicle-treated, chow-fed conventional mice increases insulin sensitivity in HFD-fed GF mice. Thus, the gut microbiota is necessary for, and can transmit, the insulin-sensitizing effects of PAHSAs in HFD-fed GF male mice. Analyses of the cecal metagenome and lipidome of PAHSA-treated mice identified multiple lipid species that associate with the gut commensal Bacteroides thetaiotaomicron (Bt) and with insulin sensitivity resulting from PAHSA treatment. Supplementing live, and to some degree, heat-killed Bt to HFD-fed female mice prevented weight gain, reduced adiposity, improved glucose tolerance, fortified the colonic mucus barrier and reduced systemic inflammation compared to HFD-fed controls. These effects were not observed in HFD-fed male mice. Furthermore, ovariectomy partially reversed the beneficial Bt effects on host metabolism, indicating a role for sex hormones in mediating the Bt probiotic effects. Altogether, these studies highlight the fact that PAHSAs can modulate the gut microbiota and that the microbiota is necessary for the beneficial metabolic effects of PAHSAs in HFD-fed mice.


Sujet(s)
Alimentation riche en graisse , Microbiome gastro-intestinal , Insulinorésistance , Obésité , Animaux , Mâle , Femelle , Souris , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Obésité/métabolisme , Obésité/microbiologie , Obésité/étiologie , Alimentation riche en graisse/effets indésirables , Souris de lignée C57BL , Acides stéariques/métabolisme , Acide palmitique/métabolisme , Fèces/microbiologie , Souris obèse
9.
Nutrients ; 16(13)2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38999751

RÉSUMÉ

To investigate the effects of rapeseed diacylglycerol oil (RDG) intake on lipid accumulation and metabolism in C57BL/6J mice, obese mice were fed a high-fat diet in which 45% of the total energy content came from RDG (RDGM group) or rapeseed triacylglycerol oil (RTGM group). This diet intervention was conducted for 12 weeks following the establishment of the obese mouse model. By the end of the experiment, the serum glucose levels of the mice in the RTGM and RDGM groups were 13.0 ± 1.3 mmol/L and 9.7 ± 1.5 mmol/L, respectively. Meanwhile, the serum triglyceride level in the RDGM group was 26.3% lower than that in the RTGM group. The weight-loss effect in the RDGM group was accompanied by a significant decrease in the white adipose tissue (WAT) index. The RDG intervention did not significantly change the antioxidant and anti-inflammatory properties of the rapeseed oil in vivo. The RDG diet improved the liver lipid metabolism abnormalities induced by a high-fat diet, leading to decreased liver damage index values (AST and ALT). Additionally, compared to that in the RTGM group, the expression of the adipogenic genes PPAR-γ and DGAT decreased in both the liver and intestine by 21.7% and 16.7% and by 38.7% and 47.2%, respectively, in the RDGM group. Further, most lipolytic genes in BAT showed no significant change after the RDG intervention. This implies that RDG regulates lipid metabolism by altering the expression of adipogenic genes in the liver, intestine, and adipose tissue, thereby reducing the accumulation of WAT. Furthermore, the RDG diet enhanced gut flora diversity, increasing the relative levels of unclassified Muribaculaceae and decreasing the levels of Dubosiella and Faecalibaculum in the mouse gut, potentially accelerating lipid metabolism. Thus, a three-month RDG diet intervention in obese mice exhibited benefits in regulating the somatotype, serum obesity-related indices, gut flora structure, and lipid metabolism in the adipose tissue, liver, and intestine.


Sujet(s)
Agents antiobésité , Alimentation riche en graisse , Diglycéride , Métabolisme lipidique , Foie , Souris de lignée C57BL , Obésité , Huile de colza , Animaux , Métabolisme lipidique/effets des médicaments et des substances chimiques , Obésité/métabolisme , Diglycéride/pharmacologie , Alimentation riche en graisse/effets indésirables , Mâle , Huile de colza/pharmacologie , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Souris , Agents antiobésité/pharmacologie , Tissu adipeux blanc/métabolisme , Tissu adipeux blanc/effets des médicaments et des substances chimiques , Triglycéride/sang , Diacylglycerol O-acyltransferase/métabolisme , Diacylglycerol O-acyltransferase/génétique , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Récepteur PPAR gamma/métabolisme , Souris obèse
10.
Nutrients ; 16(13)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38999849

RÉSUMÉ

Obesity is associated with one-fifth of cancer deaths, and breast cancer is one of the obesity-related cancers. Triple-negative breast cancer (TNBC) lacks estrogen and progesterone receptors and human epidermal growth factor receptor 2, leading to the absence of these therapeutic targets, followed by poor overall survival. We investigated if obesity could hasten TNBC progression and intermittent fasting (IF) could attenuate the progression of obesity-related TNBC. Our meta-analysis of the TNBC outcomes literature showed that obesity led to poorer overall survival in TNBC patients. Fasting-mimicking media reduced cell proliferation disrupted the cell cycle, and decreased cell migration and invasion. IF decreased body weight in obese mice but no change in normal mice. Obese mice exhibited elevated plasma glucose and cholesterol levels, increased tumor volume and weight, and enhanced macrophage accumulation in tumors. The obesity-exacerbated TNBC progression was attenuated after IF, which decreased cyclin B1 and vimentin levels and reduced the proinflammatory signature in the obesity-associated tumor microenvironment. IF attenuated obesity-induced TNBC progression through reduced obesity and tumor burdens in cell and animal experiments, supporting the potential of a cost-effective adjuvant IF therapy for TNBC through lifestyle change. Further evidence is needed of these IF benefits in TNBC, including from human clinical trials.


Sujet(s)
Cycle cellulaire , Évolution de la maladie , Transition épithélio-mésenchymateuse , Jeûne , Obésité , Tumeurs du sein triple-négatives , Animaux , Obésité/complications , Femelle , Humains , Souris , Lignée cellulaire tumorale , Inflammation , Prolifération cellulaire , Microenvironnement tumoral , Souris obèse , Mouvement cellulaire , Jeûne intermittent
11.
Int J Mol Sci ; 25(13)2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-39000013

RÉSUMÉ

Obesity is a global health concern implicated in numerous chronic degenerative diseases, including type 2 diabetes, dyslipidemia, and neurodegenerative disorders. It is characterized by chronic low-grade inflammation, gut microbiota dysbiosis, insulin resistance, glucose intolerance, and lipid metabolism disturbances. Here, we investigated the therapeutic potential of environmental enrichment (EE) to prevent the progression of gut dysbiosis in mice with high-fat diet (HFD)-induced metabolic syndrome. C57BL/6 male mice with obesity and metabolic syndrome, continuously fed with an HFD, were exposed to EE. We analyzed the gut microbiota of the mice by sequencing the 16s rRNA gene at different intervals, including on day 0 and 12 and 24 weeks after EE exposure. Fasting glucose levels, glucose tolerance, insulin resistance, food intake, weight gain, lipid profile, hepatic steatosis, and inflammatory mediators were evaluated in serum, adipose tissue, and the colon. We demonstrate that EE intervention prevents the progression of HFD-induced dysbiosis, reducing taxa associated with metabolic syndrome (Tepidimicrobium, Acidaminobacteraceae, and Fusibacter) while promoting those linked to healthy physiology (Syntrophococcus sucrumutans, Dehalobacterium, Prevotella, and Butyricimonas). Furthermore, EE enhances intestinal barrier integrity, increases mucin-producing goblet cell population, and upregulates Muc2 expression in the colon. These alterations correlate with reduced systemic lipopolysaccharide levels and attenuated colon inflammation, resulting in normalized glucose metabolism, diminished adipose tissue inflammation, reduced liver steatosis, improved lipid profiles, and a significant reduction in body weight gain despite mice's continued HFD consumption. Our findings highlight EE as a promising anti-inflammatory strategy for managing obesity-related metabolic dysregulation and suggest its potential in developing probiotics targeting EE-modulated microbial taxa.


Sujet(s)
Alimentation riche en graisse , Dysbiose , Microbiome gastro-intestinal , Souris de lignée C57BL , Obésité , Animaux , Alimentation riche en graisse/effets indésirables , Dysbiose/microbiologie , Souris , Obésité/métabolisme , Obésité/microbiologie , Mâle , Glucose/métabolisme , Souris obèse , Insulinorésistance , Syndrome métabolique X/métabolisme , Syndrome métabolique X/étiologie , Syndrome métabolique X/microbiologie
12.
J Agric Food Chem ; 72(29): 16312-16322, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-38985073

RÉSUMÉ

Sea cucumber phospholipids have ameliorative effects on various diseases related to lipid metabolism. However, it is unclear whether it can ameliorate obesity-associated glomerulopathy (ORG) induced by a high-fat diet (HFD). The present study applied UPLC-QqQ-MS/MS and atmospheric pressure matrix-assisted laser desorption ionization mass spectrometry imaging (AP-MALDI MSI) to investigate the effects of sea cucumber phospholipids, including plasmalogen PlsEtn and plasmanylcholine PakCho, on phospholipid profiles in the HFD-induced ORG mouse kidney. Quantitative analysis of 135 phospholipids revealed that PlsEtn and PakCho significantly modulated phospholipid levels. Notably, PlsEtn modulated kidney overall phospholipids better than PakCho. Imaging the "space-content" of 9 phospholipids indicated that HFD significantly increased phospholipid content within the renal cortex. Furthermore, PlsEtn and PakCho significantly decreased the expression of transport-related proteins CD36, while elevating the expression of fatty acid ß-oxidation-related protein PPAR-α in the renal cortex. In conclusion, sea cucumber phospholipids reduced renal lipid accumulation, ameliorated renal damage, effectively regulated the content and distribution of renal phospholipids, and improved phospholipid homeostasis, exerting an anti-OGR effect.


Sujet(s)
Rein , Souris de lignée C57BL , Obésité , Phospholipides , Concombres de mer , Spectrométrie de masse MALDI , Spectrométrie de masse en tandem , Animaux , Concombres de mer/composition chimique , Concombres de mer/métabolisme , Souris , Phospholipides/métabolisme , Phospholipides/composition chimique , Rein/métabolisme , Rein/composition chimique , Spectrométrie de masse en tandem/méthodes , Mâle , Spectrométrie de masse MALDI/méthodes , Chromatographie en phase liquide à haute performance/méthodes , Obésité/métabolisme , Humains , Alimentation riche en graisse/effets indésirables , Souris obèse , Maladies du rein/métabolisme
13.
Zhongguo Zhong Yao Za Zhi ; 49(13): 3600-3607, 2024 Jul.
Article de Chinois | MEDLINE | ID: mdl-39041132

RÉSUMÉ

Based on the Toll-like receptor 4(TLR4)/myeloid differentiation factor 88(MyD88)/nuclear factor kappaB(NF-κB) signaling pathway, this study observed the regulatory effect of ginsenoside Rb_1(Rb_1) on liver lipid metabolism in db/db obese mice and explored its potential mechanism. Thirty 6-week-old male db/db mice were randomly divided into a model group, a metformin group, and Rb_1 groups with low, medium, and high doses, with six mice in each group. Additionally, six age-matched male db/m mice were assigned to the normal group. The intervention lasted for five weeks. Body weight, fasting blood glucose, and food intake were mea-sured weekly. At the end of the experiment, serum lipid levels and liver function were detected. Hematoxylin-eosin(HE) staining and oil red O staining were performed to observe pathological changes in liver tissue. Real-time quantitative PCR and immunohistochemistry on paraffin sections were used to detect the mRNA and protein expression of TLR4, MyD88, and NF-κB p65. RESULTS:: showed that compared with the normal group, the model group exhibited significant increases in body weight, liver weight, liver index, epididymal fat mass, epididymal fat index, total cholesterol, low-density lipoprotein cholesterol, liver function parameters, and fasting blood glucose levels. Liver lipid accumulation significantly increased, along with elevated mRNA and protein expression of TLR4, MyD88, and NF-κB p65 in the liver. After Rb_1 treatment, the above-mentioned parameters in the intervention groups showed significant reversals. In conclusion, Rb_1 can improve obesity and obesity-related hepatic steatosis in mice while regulating abnormal lipid and glucose meta-bolism. Mechanistically, Rb_1 may improve liver steatosis in db/db obese mice by modulating the TLR4/MyD88/NF-κB signaling pathway.


Sujet(s)
Stéatose hépatique , Ginsénosides , Facteur de différenciation myéloïde-88 , Facteur de transcription NF-kappa B , Transduction du signal , Récepteur de type Toll-4 , Animaux , Ginsénosides/pharmacologie , Ginsénosides/administration et posologie , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/métabolisme , Facteur de différenciation myéloïde-88/génétique , Facteur de différenciation myéloïde-88/métabolisme , Souris , Mâle , Facteur de transcription NF-kappa B/génétique , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Stéatose hépatique/traitement médicamenteux , Stéatose hépatique/métabolisme , Stéatose hépatique/génétique , Obésité/traitement médicamenteux , Obésité/métabolisme , Obésité/génétique , Souris obèse , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Humains , Médicaments issus de plantes chinoises/administration et posologie , Médicaments issus de plantes chinoises/pharmacologie
14.
J Med Chem ; 67(14): 11814-11826, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38977267

RÉSUMÉ

Peptide-based drug discovery has surged with the development of peptide hormone-derived analogs for the treatment of diabetes and obesity. Machine learning (ML)-enabled quantitative structure-activity relationship (QSAR) approaches have shown great promise in small molecule drug discovery but have been less successful in peptide drug discovery due to limited data availability. We have developed a peptide drug discovery platform called streaMLine, enabling rigorous design, synthesis, screening, and ML-driven analysis of large peptide libraries. Using streaMLine, this study systematically explored secretin as a peptide backbone to generate potent, selective, and long-acting GLP-1R agonists with improved physicochemical properties. We synthesized and screened a total of 2688 peptides and applied ML-guided QSAR to identify multiple options for designing stable and potent GLP-1R agonists. One candidate, GUB021794, was profiled in vivo (S.C., 10 nmol/kg QD) and showed potent body weight loss in diet-induced obese mice and a half-life compatible with once-weekly dosing.


Sujet(s)
Découverte de médicament , Récepteur du peptide-1 similaire au glucagon , Apprentissage machine , Récepteur du peptide-1 similaire au glucagon/agonistes , Animaux , Souris , Humains , Peptides/composition chimique , Peptides/pharmacologie , Peptides/synthèse chimique , Obésité/traitement médicamenteux , Souris de lignée C57BL , Mâle , Relation quantitative structure-activité , Souris obèse ,
15.
Mol Metab ; 86: 101979, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38945296

RÉSUMÉ

OBJECTIVE: Bariatric surgery is an effective treatment to obesity, leading to weight loss and improvement in glycemia, that is characterized by hypersecretion of gastrointestinal hormones. However, weight regain and relapse of hyperglycemia are not uncommon. We set to identify mechanisms that can enhance gastrointestinal hormonal secretion following surgery to sustain weight loss. METHODS: We investigated the effect of somatostatin (Sst) inhibition on the outcomes of bariatric surgery using a mouse model of sleeve gastrectomy (SG). RESULTS: Sst knockout (sst-ko) mice fed with a calorie-rich diet gained weight normally and had a mild favorable metabolic phenotype compared to heterozygous sibling controls, including elevated plasma levels of GLP-1. Mathematical modeling of the feedback inhibition between Sst and GLP-1 showed that Sst exerts its maximal effect on GLP-1 under conditions of high hormonal stimulation, such as following SG. Obese sst-ko mice that underwent SG had higher levels of GLP-1 compared with heterozygous SG-operated controls. The SG-sst-ko mice regained less weight than controls and maintained lower glycemia months after surgery. Obese wild-type mice that underwent SG and were treated daily with a Sst receptor inhibitor for two months had higher GLP-1 levels, regained less weight, and improved metabolic profile compared to saline-treated SG-operated controls, and compared to inhibitor or saline-treated sham-operated obese mice. CONCLUSIONS: Our results suggest that inhibition of Sst signaling enhances the long-term favorable metabolic outcomes of bariatric surgery.


Sujet(s)
Gastrectomie , Glucagon-like peptide 1 , Souris knockout , Obésité , Somatostatine , Animaux , Somatostatine/métabolisme , Souris , Gastrectomie/méthodes , Glucagon-like peptide 1/métabolisme , Glucagon-like peptide 1/sang , Obésité/métabolisme , Obésité/chirurgie , Mâle , Chirurgie bariatrique/méthodes , Souris de lignée C57BL , Perte de poids , Glycémie/métabolisme , Souris obèse
16.
Eur J Pharmacol ; 978: 176789, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-38945287

RÉSUMÉ

The increased incidence of obesity, which become a global health problem, requires more functional food products with minor side and excellent effects. Calebin A (CbA) is a non-curcuminoid compound, which is reported to be an effective treatment for lipid metabolism and thermogenesis. However, its ability and mechanism of action in improving obesity-associated hyperglycemia remain unclear. This study was designed to explore the effect and mechanism of CbA in hyperglycemia via improvement of inflammation and glucose metabolism in the adipose tissue and liver in high-fat diet (HFD)-fed mice. After 10 weeks fed HFD, obese mice supplemented with CbA (25 and 100 mg/kg) for another 10 weeks showed a remarkable reducing adiposity and blood glucose. CbA modulated M1/M2 macrophage polarization, ameliorated inflammatory cytokines, and restored adiponectin as well as Glut 4 expression in the adipose tissue. In the in vitro study, CbA attenuated pro-inflammatory markers while upregulated anti-inflammatory IL-10 in LPS + IFNγ-generated M1 phenotype macrophages. In the liver, CbA attenuated steatosis, inflammatory infiltration, and protein levels of inflammatory TNF-α and IL-6. Moreover, CbA markedly upregulated Adiponectin receptor 1, AMPK, and insulin downstream Akt signaling to improve glycogen content and increase Glut2 protein. These findings indicated that CbA may be a novel therapeutic approach to treat obesity and hyperglycemia phenotype targeting on adipose inflammation and hepatic insulin signaling.


Sujet(s)
Tissu adipeux , Alimentation riche en graisse , Glucose , Hyperglycémie , Inflammation , Foie , Macrophages , Obésité , Animaux , Alimentation riche en graisse/effets indésirables , Souris , Hyperglycémie/traitement médicamenteux , Hyperglycémie/métabolisme , Tissu adipeux/effets des médicaments et des substances chimiques , Tissu adipeux/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Mâle , Glucose/métabolisme , Obésité/traitement médicamenteux , Obésité/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/anatomopathologie , Cellules RAW 264.7 , Souris obèse , Souris de lignée C57BL , Transduction du signal/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Glycémie/métabolisme , Glycémie/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme
17.
Biomed Pharmacother ; 177: 116926, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38906016

RÉSUMÉ

Obesity aggravates ferroptosis, and vitamin D (VD) may inhibit ferroptosis. We hypothesized that weight reduction and/or calcitriol administration have benefits against the sepsis-induced liver redox imbalance and ferroptosis in obese mice. Mice were fed a high-fat diet for 11 weeks, then half of the mice continued to consume the diet, while the other half were transferred to a low-energy diet for 5 weeks. After feeding the respective diets for 16 weeks, sepsis was induced by cecal ligation and puncture (CLP). Septic mice were divided into four experimental groups: OS group, obese mice injected with saline; OD group, obese mice with calcitriol; WS group, weight-reduction mice with saline; and WD group, weight-reduction mice with calcitriol. Mice in the respective groups were euthanized at 12 or 24 h after CLP. Results showed that the OS group had the highest inflammatory mediators and lipid peroxide levels in the liver. Calcitriol treatment reduced iron content, enhanced the reduced glutathione/oxidized glutathione ratio, upregulated nuclear factor erythroid 2-related factor 2, ferroptosis-suppressing protein 1, and solute carrier family 7 member 11 expression levels. Also, mitochondrion-associated nicotinamide adenine dinucleotide phosphate oxidase 1, peroxisome proliferator-activated receptor-γ coactivator 1, hypoxia-inducible factor-1α, and heme oxidase-1 expression levels increased in the late phase of sepsis. These results were not noted in the WS group. These findings suggest that calcitriol treatment elicits a more-balanced glutathione redox status, alleviates liver ferroptosis, and enhances mitochondrial biogenesis-associated gene expressions. Weight reduction alone had minimal influences on liver ferroptosis and mitochondrial biogenesis in obese mice with sepsis.


Sujet(s)
Calcitriol , Alimentation riche en graisse , Ferroptose , Foie , Souris de lignée C57BL , Obésité , Oxydoréduction , Sepsie , Animaux , Sepsie/complications , Sepsie/traitement médicamenteux , Sepsie/métabolisme , Alimentation riche en graisse/effets indésirables , Obésité/métabolisme , Obésité/complications , Obésité/traitement médicamenteux , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Oxydoréduction/effets des médicaments et des substances chimiques , Ferroptose/effets des médicaments et des substances chimiques , Mâle , Souris , Calcitriol/pharmacologie , Calcitriol/administration et posologie , Souris obèse
18.
Gene ; 927: 148604, 2024 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-38838872

RÉSUMÉ

OBJECTIVE: To extract exosomes from obese and non-obese mice, screen specifically expressed microRNAs by high-throughput sequencing and explore their roles. METHODS: An animal obesity model was constructed, and the successful construction of the obesity model was verified by HE staining, Western Blot and RT-qPCR. In addition, exosomes were extracted and verified by Western Blot. High-throughput sequencing was performed on the extracted serum exosomes to screen for differentially expressed microRNAs. fluorescence quantitative RT-PCR (RT-qPCR) was used to validate the differentially expressed miRNAs and explore their functions. RESULTS: 8 microRNAs were up-regulated and 11 microRNAs were down-regulated. mmu-miR-674-5p and X_28316 were significantly down-regulated and had the greatest impact on protein pathways. 8_13258 was significantly up-regulated and affected multiple protein pathways. GO enrichment analysis suggested that the differentially expressed microRNAs were mainly involved in the cleavage of microtubule activity, transferase activity/transferase pentameric acid. GO enrichment analysis suggested that differentially expressed microRNAs were mainly involved in the processes of cleavage microtubule activity, transferase activity/transfer pentamer, and threonine phosphatase/threonine kinase activity.KEGG pathway enrichment analysis showed that differentially expressed microRNAs were mainly involved in the processes of regulating the phosphorylation of TP53 activity, the G2/M DNA damage checkpoint, and the processing of the ends of DNA double-strand breaks. Protein interaction networks were enriched for Stat3, Fgr, Camk2b, Rac1, Asb6, and Ankfy1. Suggesting that they may be mediated by differential genes to participate in the process of insulin resistance. qRT-PCR results showed that the expression trend of mmu-miR-674-5p was consistent with the sequencing results. It suggests that it may be able to participate in the regulation of insulin resistance as a target gene. CONCLUSION: microRNAs were differentially expressed in serum exosomes of obese and non-obese mice and might be involved in the specific regulation of insulin resistance. mmu-miR-674-5p was differentially expressed significantly and the validation trend was consistent with it, suggesting that it might be able to participate in the regulation of insulin resistance as a target gene.


Sujet(s)
Exosomes , microARN , Obésité , Animaux , Exosomes/métabolisme , Exosomes/génétique , Souris , microARN/génétique , microARN/sang , Obésité/génétique , Obésité/sang , Obésité/métabolisme , Mâle , Séquençage nucléotidique à haut débit , Souris obèse , Analyse de profil d'expression de gènes/méthodes , Souris de lignée C57BL , Régulation de l'expression des gènes , Modèles animaux de maladie humaine
19.
Microbiome ; 12(1): 103, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38845049

RÉSUMÉ

BACKGROUND: The metabolic disturbances of obesity can be mitigated by strategies modulating the gut microbiota. In this study, we sought to identify whether innate or adaptive immunity mediates the beneficial metabolic effects of the human intestinal bacterium Bacteroides uniformis CECT 7771 in obesity. METHODS: We evaluated the effects of orally administered B. uniformis on energy homeostasis, intestinal immunity, hormone levels, and gut microbiota in wild-type and Rag1-deficient mice with diet-induced obesity. We also assessed whether B. uniformis needed to be viable to exert its beneficial effects in obesity and to directly induce immunoregulatory effects. RESULTS: The administration of B. uniformis to obese mice improved glucose tolerance and insulin secretion, restored the caloric intake suppression after an oral glucose challenge, and reduced hyperglycemia. The pre- and post-prandial glucose-related benefits were associated with restoration of the anti-inflammatory tone mediated by type 2 macrophages and regulatory T cells (Tregs) in the lamina propria of the small intestine. Contrastingly, B. uniformis administration failed to improve glucose tolerance in obese Rag1-/- mice, but prevented the increased body weight gain and adiposity. Overall, the beneficial effects seemed to be independent of enteroendocrine effects and of major changes in gut microbiota composition. B. uniformis directly induced Tregs generation from naïve CD4+ T cells in vitro and was not required to be viable to improve glucose homeostasis but its viability was necessary to prevent body weight gain in diet-induced obese wild-type mice. CONCLUSIONS: Here we demonstrate that B. uniformis modulates the energy homeostasis in diet-induced obese mice through different mechanisms. The bacterium improves oral glucose tolerance by adaptive immunity-dependent mechanisms that do not require cell viability and prevents body weight gain by adaptive immunity-independent mechanisms which require cell viability. Video Abstract.


Sujet(s)
Immunité acquise , Bacteroides , Microbiome gastro-intestinal , Obésité , Prise de poids , Animaux , Souris , Obésité/immunologie , Obésité/microbiologie , Alimentation riche en graisse/effets indésirables , Souris obèse , Lymphocytes T régulateurs/immunologie , Souris de lignée C57BL , Mâle , Humains , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Probiotiques/administration et posologie , Souris knockout , Glucose/métabolisme
20.
Nat Commun ; 15(1): 4704, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38830845

RÉSUMÉ

Metabolic syndrome encompasses amongst other conditions like obesity and type-2 diabetes and is associated with gut microbiome (GM) dysbiosis. Fecal microbiota transplantation (FMT) has been explored to treat metabolic syndrome by restoring the GM; however, concerns on accidentally transferring pathogenic microbes remain. As a safer alternative, fecal virome transplantation (FVT, sterile-filtrated feces) has the advantage over FMT in that mainly bacteriophages are transferred. FVT from lean male donors have shown promise in alleviating the metabolic effects of high-fat diet in a preclinical mouse study. However, FVT still carries the risk of eukaryotic viral infections. To address this, recently developed methods are applied for removing or inactivating eukaryotic viruses in the viral component of FVT. Modified FVTs are compared with unmodified FVT and saline in a diet-induced obesity model on male C57BL/6 N mice. Contrasted with obese control, mice administered a modified FVT (nearly depleted for eukaryotic viruses) exhibits enhanced blood glucose clearance but not weight loss. The unmodified FVT improves liver pathology and reduces the proportions of immune cells in the adipose tissue with a non-uniform response. GM analysis suggests that bacteriophage-mediated GM modulation influences outcomes. Optimizing these approaches could lead to the development of safe bacteriophage-based therapies targeting metabolic syndrome through GM restoration.


Sujet(s)
Alimentation riche en graisse , Transplantation de microbiote fécal , Microbiome gastro-intestinal , Syndrome métabolique X , Souris de lignée C57BL , Souris obèse , Obésité , Virome , Animaux , Mâle , Syndrome métabolique X/thérapie , Obésité/thérapie , Souris , Alimentation riche en graisse/effets indésirables , Dysbiose/thérapie , Fèces/virologie , Fèces/microbiologie , Bactériophages/physiologie , Glycémie/métabolisme , Modèles animaux de maladie humaine , Foie/anatomopathologie , Foie/métabolisme , Tissu adipeux
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE