Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.860
Filtrer
1.
Nutrients ; 16(15)2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39125345

RÉSUMÉ

In this study, we undertook an extensive investigation to determine how CypB PPIase activity affects preadipocyte differentiation and lipid metabolism. Our findings revealed that inhibition of CypB's PPIase activity suppressed the expression of crucial proteins involved in adipocyte differentiation and induced changes in proteins regulating the cell cycle. Furthermore, we clarified the impact of CypB's PPIase activity on lipid metabolism via the AKT/mTOR signaling pathway. Additionally, we demonstrated the involvement of CypB's PPIase activity in lipid metabolism through the XBP1s pathway. These discoveries offer invaluable insights for devising innovative therapeutic strategies aimed at treating and averting obesity and its related health complications. Targeting CypB's PPIase activity may emerge as a promising avenue for addressing obesity-related conditions. Furthermore, our research opens up opportunities for creating new therapeutic strategies by enhancing our comprehension of the processes involved in cellular endoplasmic reticulum stress.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Différenciation cellulaire , Métabolisme lipidique , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Protéine-1 liant la boite X , Protéine-1 liant la boite X/métabolisme , Animaux , Souris , Sérine-thréonine kinases TOR/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Adipocytes/métabolisme , Adipogenèse , Stress du réticulum endoplasmique/physiologie
2.
Int J Mol Sci ; 25(15)2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-39125811

RÉSUMÉ

Advanced glycated end products (AGEs) are cytotoxic compounds that are mainly increased in diabetes mellitus (DM), kidney failure, inflammation, and in response to the ingestion of AGE-rich diets. AGEs can also impair glycemic homeostasis by decreasing the expression of the Slc2a4 (solute carrier family 2 member 4) gene and its GLUT4 (solute carrier family 2, facilitated glucose transporter member 4) protein in muscle. However, the mechanisms underlying AGE's effect on adipocytes have not been demonstrated yet. This study investigated the effects of AGEs upon Slc2a4/GLUT4 expression in 3T3-L1 adipocytes, as well as the potential role of NFKB (nuclear factor NF-kappa-B) activity in the effects observed. Adipocytes were cultured in the presence of control albumin (CA) or advanced glycated albumin (GA) at concentrations of 0.4, 3.6, and 5.4 mg/mL for 24 h or 72 h. Slc2a4, Rela, and Nfkb1mRNAs were measured by RT-qPCR, GLUT4, IKKA/B, and p50/p65 NFKB subunits using Western blotting, and p50/p65 binding into the Slc2a4 promoter was analyzed by chromatin immunoprecipitation (ChIP) assay. GA at 0.4 mg/mL increased Slc2a4/GLUT4 expression after 24 h and 72 h (from 50% to 100%), but at 5.4 mg/mL, Slc2a4/GLUT4 expression decreased at 72 h (by 50%). Rela and Nfkb1 expression increased after 24 h at all concentrations, but this effect was not observed at 72 h. Furthermore, 5.4 mg/mL of GA increased the p50/p65 nuclear content and binding into Slc2a4 at 72 h. In summary, this study reveals AGE-induced and NFKB-mediated repression of Slc2a4/GLUT4 expression. This can compromise the adipocyte glucose utilization, contributing not only to the worsening of glycemic control in DM subjects but also the impairment of glycemic homeostasis in non-DM subjects under the high intake of AGE-rich foods.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Transporteur de glucose de type 4 , Produits terminaux de glycation avancée , Facteur de transcription RelA , Animaux , Transporteur de glucose de type 4/métabolisme , Transporteur de glucose de type 4/génétique , Souris , Produits terminaux de glycation avancée/métabolisme , Produits terminaux de glycation avancée/pharmacologie , Adipocytes/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Facteur de transcription RelA/métabolisme , Facteur de transcription RelA/génétique , Facteur de transcription NF-kappa B/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Régions promotrices (génétique) , Sous-unité p50 de NF-kappa B/métabolisme , Sous-unité p50 de NF-kappa B/génétique
3.
PLoS One ; 19(8): e0306903, 2024.
Article de Anglais | MEDLINE | ID: mdl-39116155

RÉSUMÉ

Malva parviflora has shown anti-inflammatory, antihypertensive, antihyperlipidemic, and hypoglycemic effects. This study is aimed to evaluate the anti-adipogenic effect of M. parviflora on 3T3-L1 adipocytes. Fibroblast differentiation was induced either in the absence or presence of M. parviflora fractions (F3, F4, F7, F12, F13, F17, F18 and F19) for 4 days; F17 and 18 were the most effective fractions in reducing intracellular lipid accumulation (by 25.6% and 23.1%, respectively). EC50 of F17 and F18 (14 µg/mL and 17 µg/mL, respectively) were used to evaluate their anti adipogenic effect. After 10 days of inducing differentiation in the absence or presence of the extracts at the EC50 of F17 and F18, lipid accumulation, the concentration of interleukin 6 (IL-6) were measured in the culture medium; the presence of PPAR-γ, AKT, and p-AKT was also determined. In differentiated adipocytes (C2), F17 maintained intracellular lipid concentration at levels comparable to metformin, while decreasing PPAR-γ and increasing p-AKT presence; it also prevented IL-6 expression. F17 consists of alanine, valine, phenylalanine, and proline. On the other hand, F18 reduced intracellular lipid concentrations, prevented the increase of PPAR-γ and p-AKT, and maintained IL-6 expression at similar levels as metformin. F18 is mainly constituted by alanine, valine, proline, and sucrose. In conclusion, M. parviflora fractions (F17 and F18) control the process of adipogenesis, lipogenesis, and cellular dysfunction.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Adipogenèse , Récepteur PPAR gamma , Extraits de plantes , Animaux , Souris , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Adipocytes/cytologie , Adipogenèse/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Récepteur PPAR gamma/métabolisme , Interleukine-6/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Métabolisme lipidique/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme
4.
Signal Transduct Target Ther ; 9(1): 218, 2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39174522

RÉSUMÉ

Obesity is a global issue that warrants the identification of more effective therapeutic targets and a better understanding of the pivotal molecular pathogenesis. Annexin A1 (ANXA1) is known to inhibit phospholipase A2, exhibiting anti-inflammatory activity. However, the specific effects of ANXA1 in obesity and the underlying mechanisms of action remain unclear. Our study reveals that ANXA1 levels are elevated in the adipose tissue of individuals with obesity. Whole-body or adipocyte-specific ANXA1 deletion aggravates obesity and metabolic disorders. ANXA1 levels are higher in stromal vascular fractions (SVFs) than in mature adipocytes. Further investigation into the role of ANXA1 in SVFs reveals that ANXA1 overexpression induces lower numbers of mature adipocytes, while ANXA1-knockout SVFs exhibit the opposite effect. This suggests that ANXA1 plays an important role in adipogenesis. Mechanistically, ANXA1 competes with MYC binding protein 2 (MYCBP2) for interaction with PDZ and LIM domain 7 (PDLIM7). This exposes the MYCBP2-binding site, allowing it to bind more readily to the SMAD family member 4 (SMAD4) and promoting its ubiquitination and degradation. SMAD4 degradation downregulates peroxisome proliferator-activated receptor gamma (PPARγ) transcription and reduces adipogenesis. Treatment with Ac2-26, an active peptide derived from ANXA1, inhibits both adipogenesis and obesity through the mechanism. In conclusion, the molecular mechanism of ANXA1 inhibiting adipogenesis was first uncovered in our study, which is a potential target for obesity prevention and treatment.


Sujet(s)
Adipocytes , Adipogenèse , Annexine A1 , Obésité , Récepteur PPAR gamma , Annexine A1/génétique , Annexine A1/métabolisme , Adipogenèse/génétique , Animaux , Obésité/génétique , Obésité/métabolisme , Obésité/anatomopathologie , Humains , Souris , Récepteur PPAR gamma/génétique , Récepteur PPAR gamma/métabolisme , Adipocytes/métabolisme , Adipocytes/anatomopathologie , Protéine Smad-4/génétique , Protéine Smad-4/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Cellules 3T3-L1 , Peptides
5.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-39062809

RÉSUMÉ

The study evaluated the effects of Arthrospira maxima phycobiliproteins (PBPs), rosiglitazone (RSG), and 17ß-estradiol (E) on the differentiation process of 3T3-L1 cells and on their regulation of lipogenic and inflammatory gene expression at different stages of the process. The results showed that phycobiliproteins promoted cell proliferation after 24 h of treatment. Furthermore, for all three treatments, the regulation of the highest number of markers occurred on days 6 and 12 of differentiation, regardless of when the treatment was applied. Phycobiliproteins reduced lipid droplet accumulation on days 3, 6, 10, and 13 of the adipogenic process, while rosiglitazone showed no differences compared to the control. On day 6, both phycobiliproteins and rosiglitazone positively regulated Acc1 mRNA. Meanwhile, all three treatments negatively regulated Pparγ and C/ebpα. Phycobiliproteins and estradiol also negatively regulated Ucp1 and Glut4 mRNAs. Rosiglitazone and estradiol, on the other hand, negatively regulated Ppara and Il-6 mRNAs. By day 12, phycobiliproteins and rosiglitazone upregulated Pparγ mRNA and negatively regulated Tnfα and Il-1ß. Additionally, phycobiliproteins and estradiol positively regulated Il-6 and negatively regulated Ppara, Ucp2, Acc1, and Glut4. Rosiglitazone and estradiol upregulate C/ebpα and Ucp1 mRNAs. The regulation exerted by phycobiliproteins on the mRNA expression of the studied markers was dependent on the phase of cell differentiation. The results of this study highlight that phycobiliproteins have an anti-adipogenic and anti-inflammatory effect by reducing the expression of adipogenic, lipogenic, and inflammatory genes in 3T3-L1 cells at different stages of the differentiation process.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Adipogenèse , Différenciation cellulaire , Oestradiol , Phycobiliprotéines , Rosiglitazone , Animaux , Souris , Oestradiol/pharmacologie , Rosiglitazone/pharmacologie , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Adipocytes/cytologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Adipogenèse/effets des médicaments et des substances chimiques , Adipogenèse/génétique , Phycobiliprotéines/pharmacologie , Phycobiliprotéines/métabolisme , Phycobiliprotéines/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Lipogenèse/effets des médicaments et des substances chimiques , Lipogenèse/génétique , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/génétique , Spirulina
6.
Sci Rep ; 14(1): 15064, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956395

RÉSUMÉ

Sargassum horneri (S. horneri), a brown seaweed excessively proliferating along Asian coastlines, are damaging marine ecosystems. Thus, this study aimed to enhance nutritional value of S. horneri through lactic acid bacteria fermentation to increase S. horneri utilization as a functional food supplement, and consequently resolve coastal S. horneri accumulation. S. horneri supplemented fermentation was most effective with Lactiplantibacillus pentosus SH803, thus this product (F-SHWE) was used for further in vitro studies. F-SHWE normalized expressions of oxidative stress related genes NF-κB, p53, BAX, cytochrome C, caspase 9, and caspase 3, while non-fermented S. horneri (SHWE) did not, in a H2O2-induced HT-29 cell model. Moreover, in an LPS-induced HT-29 cell model, F-SHWE repaired expressions of inflammation marker genes ZO1, IL1ß, IFNγ more effectively than SHWE. For further functional assessment, F-SHWE was also treated in 3T3-L1 adipocytes. As a result, F-SHWE decreased lipid accumulation, along with gene expression of adipogenesis markers PPARγ, C/EBPα, C/EBPß, aP2, and Lpl; lipogenesis markers Lep, Akt, SREBP1, Acc, Fas; inflammation markers IFN-γ and NF-κB. Notably, gene expression of C/EBPß, IFN-γ and NF-κB were suppressed only by F-SHWE, suggesting the enhancing effect of fermentation on obesity-related properties. Compositional analysis attributed the protective effects of F-SHWE to acetate, an organic acid significantly higher in F-SHWE than SHWE. Therefore, F-SHWE is a novel potential anti-obesity agent, providing a strategy to reduce excess S. horneri populations along marine ecosystems.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Fermentation , Inflammation , Stress oxydatif , Sargassum , Sargassum/composition chimique , Souris , Animaux , Adipocytes/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Humains , Inflammation/métabolisme , Lactobacillus pentosus/métabolisme , Cellules HT29 , Adipogenèse/effets des médicaments et des substances chimiques
7.
Nutrients ; 16(13)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38999918

RÉSUMÉ

This study aimed to investigate the therapeutic potential of Citrullus mucosospermus extract (CME) in counteracting adipogenesis and its associated metabolic disturbances in murine models. In vitro experiments utilizing 3T3-L1 preadipocytes revealed that CME potently inhibited adipocyte differentiation, as evidenced by a dose-dependent reduction in lipid droplet formation. Remarkably, CME also attenuated glucose uptake and intracellular triglyceride accumulation in fully differentiated adipocytes, suggesting its ability to modulate metabolic pathways in mature adipose cells. Translating these findings to an in vivo setting, we evaluated the effects of CME in C57BL/6N mice fed a high-fat diet (HFD) for 10 weeks. CME administration, concomitantly with the HFD, resulted in a significant attenuation of body weight gain compared to the HFD control group. Furthermore, CME treatment led to substantial reductions in liver weight, total fat mass, and deposits of visceral and retroperitoneal adipose tissue, underscoring its targeted impact on adipose expansion. Histological analyses revealed the remarkable effects of CME on hepatic steatosis. While the HFD group exhibited severe lipid accumulation within liver lobules, CME dose-dependently mitigated this pathology, with the highest dose virtually abolishing hepatic fat deposition. An examination of adipose tissue revealed a progressive reduction in adipocyte hypertrophy upon CME treatment, culminating in a near-normalization of adipocyte morphology at the highest dose. Notably, CME exhibited potent anti-inflammatory properties, significantly attenuating the upregulation of pro-inflammatory cytokines' mRNA levels (TNF-α, IL-1ß and IL-6) in the livers of HFD-fed mice. This suggests a potential mechanism through which CME may exert protective effects against inflammation associated with obesity and fatty liver disease.


Sujet(s)
Cellules 3T3-L1 , Adipogenèse , Alimentation riche en graisse , Souris de lignée C57BL , Extraits de plantes , Prise de poids , Animaux , Alimentation riche en graisse/effets indésirables , Extraits de plantes/pharmacologie , Souris , Prise de poids/effets des médicaments et des substances chimiques , Mâle , Adipogenèse/effets des médicaments et des substances chimiques , Adipocytes/effets des médicaments et des substances chimiques , Obésité , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Tissu adipeux/effets des médicaments et des substances chimiques , Tissu adipeux/métabolisme
8.
Phytomedicine ; 132: 155854, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39032276

RÉSUMÉ

BACKGROUND: Recent investigations underscore the capacity of photodynamic therapy (PDT) to induce adipocyte apoptosis, thereby mitigating obesity. Nonetheless, extant synthetic photosensitizers manifest limitations that hinder their clinical viability. PURPOSE: In the current study, we used Hypericum perforatum-derived exosomes-like nanovesicles (HPExos) as a novel photosensitizer, and investigated its PDT effects in adipose tissue during obesity. METHOD: HPExos-were administered to high fat diet mice via intraperitoneal injection, followed by targeted irradiation with specialized LED lights. Mass spectrometric analysis was analyzed in adipose tissues. CCK8 assay and Oil Red O staining were used to investigate lipid accumulation in 3T3-L1 cells to clarify adipocyte differentiation. The expression levels of related markers associated with adipogenesis and lipogenesis were assessed by RT-PCR. Apoptosis analysis was performed by TUNEL staining of and western blotting. RESULTS: HPExos combined with PDT accumulated in visceral white adipose tissues results in a reduced body weight and improved insulin sensitivity. HPExos combined with PDT induced apoptosis by driving high levels of ROS. In addition, HPExos combined with PDT significantly downregulated the expression of transcription factors, PPARγ, C/EBPα, and SREBP and lipogenesis protein FABP4 both in vitro and in vivo, associated with a decreased FFA levels. CONCLUSION: These findings suggest that HPExos could act as an effective photosensitizer in regulating glucose hemostasis by inhibiting adipocyte differentiation and lipogenesis, offering a promising approach for obesity treatment.


Sujet(s)
Cellules 3T3-L1 , Apoptose , Exosomes , Hypericum , Obésité , Photothérapie dynamique , Hypericum/composition chimique , Animaux , Souris , Exosomes/métabolisme , Photothérapie dynamique/méthodes , Mâle , Apoptose/effets des médicaments et des substances chimiques , Obésité/traitement médicamenteux , Alimentation riche en graisse , Souris de lignée C57BL , Tissu adipeux/effets des médicaments et des substances chimiques , Photosensibilisants/pharmacologie , Récepteur PPAR gamma/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Adipogenèse/effets des médicaments et des substances chimiques , Protéine alpha liant les séquences stimulatrices de type CCAAT/métabolisme , Lipogenèse/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Extraits de plantes/pharmacologie , Insulinorésistance , Protéines liant les séquences stimulatrices de type CCAAT , Protéines de liaison aux acides gras , Protéine-1 de liaison à l'élément de régulation des stérols
9.
Clin Epigenetics ; 16(1): 96, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-39033117

RÉSUMÉ

BACKGROUND: Obesity is a major health burden. Preadipocytes proliferate and differentiate in mature adipocytes in the adipogenic process, which could be a potential therapeutic approach for obesity. Deficiency of SIRT6, a stress-responsive protein deacetylase and mono-ADP ribosyltransferase enzyme, blocks adipogenesis. Mutants of SIRT6 (N308K/A313S) were recently linked to the in the long lifespan Ashkenazi Jews. In this study, we aimed to clarify how these new centenarian-associated SIRT6 genetic variants affect adipogenesis at the transcriptional and epigenetic level. METHODS: We analyzed the role of SIRT6 wild-type (WT) or SIRT6 centenarian-associated mutant (N308K/A313S) overexpression in adipogenesis, by creating stably transduced preadipocyte cell lines using lentivirus on the 3T3-L1 model. Histone post-translational modifications (PTM: acetylation, methylation) and transcriptomic changes were analyzed by mass spectrometry (LC-MS/MS) and RNA-Seq, respectively, in 3T3-L1 adipocytes. In addition, the adipogenic process and related signaling pathways were investigated by bioinformatics and biochemical approaches. RESULTS: Overexpression of centenarian-associated SIRT6 mutant increased adipogenic differentiation to a similar extent compared to the WT form. However, it triggered distinct histone PTM profiles in mature adipocytes, with significantly higher acetylation levels, and activated divergent transcriptional programs, including those dependent on signaling related to the sympathetic innervation and to PI3K pathway. 3T3-L1 mature adipocytes overexpressing SIRT6 N308K/A313S displayed increased insulin sensitivity in a neuropeptide Y (NPY)-dependent manner. CONCLUSIONS: SIRT6 N308K/A313S overexpression in mature adipocytes ameliorated glucose sensitivity and impacted sympathetic innervation signaling. These findings highlight the importance of targeting SIRT6 enzymatic activities to regulate the co-morbidities associated with obesity.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Adipogenèse , Épigenèse génétique , Sirtuines , Sirtuines/génétique , Sirtuines/métabolisme , Souris , Adipocytes/métabolisme , Animaux , Épigenèse génétique/génétique , Adipogenèse/génétique , Humains , Mutation , Obésité/génétique , Obésité/métabolisme , Maturation post-traductionnelle des protéines/génétique , Histone/métabolisme , Histone/génétique
10.
Adipocyte ; 13(1): 2374062, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38953241

RÉSUMÉ

Obesity is associated with a low-grade chronic inflammatory process characterized by higher circulating TNFα levels, thus contributing to insulin resistance. This study evaluated the effect of silybin, the main bioactive component of silymarin, which has anti-inflammatory properties, on TNFα levels and its impact on glucose uptake in the adipocyte cell line 3T3-L1 challenged with two different inflammatory stimuli, TNFα or lipopolysaccharide (LPS). Silybin's pre-treatment effect was evaluated in adipocytes pre-incubated with silybin (30 or 80 µM) before challenging with the inflammatory stimuli (TNFα or LPS). For the post-treatment effect, the adipocytes were first challenged with the inflammatory stimuli and then post-treated with silybin. After treatments, TNFα production, glucose uptake, and GLUT4 protein expression were determined. Both inflammatory stimuli increased TNFα secretion, diminished GLUT4 expression, and significantly decreased glucose uptake. Silybin 30 µM only reduced TNFα secretion after the LPS challenge. Silybin 80 µM as post-treatment or pre-treatment decreased TNFα levels, improving glucose uptake. However, glucose uptake enhancement induced by silybin did not depend on GLUT4 protein expression. These results show that silybin importantly reduced TNFα levels and upregulates glucose uptake, independently of GLUT4 protein expression.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Glucose , Lipopolysaccharides , Silibinine , Facteur de nécrose tumorale alpha , Animaux , Silibinine/pharmacologie , Souris , Facteur de nécrose tumorale alpha/métabolisme , Glucose/métabolisme , Adipocytes/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Lipopolysaccharides/pharmacologie , Transporteur de glucose de type 4/métabolisme , Silymarine/pharmacologie
11.
Cell Rep ; 43(7): 114491, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39002127

RÉSUMÉ

Tissues release microRNAs (miRNAs) in small extracellular vesicles (sEVs) including exosomes, which can regulate gene expression in distal cells, thus acting as modulators of local and systemic metabolism. Here, we show that insulin regulates miRNA secretion into sEVs from 3T3-L1 adipocytes and that this process is differentially regulated from cellular expression. Thus, of the 53 miRNAs upregulated and 66 miRNAs downregulated by insulin in 3T3-L1 sEVs, only 12 were regulated in parallel in cells. Insulin regulated this process in part by phosphorylating hnRNPA1, causing it to bind to AU-rich motifs in miRNAs, mediating their secretion into sEVs. Importantly, 43% of insulin-regulated sEV-miRNAs are implicated in obesity and insulin resistance. These include let-7 and miR-103, which we show regulate insulin signaling in AML12 hepatocytes. Together, these findings demonstrate an important layer to insulin's regulation of adipose biology and provide a mechanism of tissue crosstalk in obesity and other hyperinsulinemic states.


Sujet(s)
Vésicules extracellulaires , Insuline , microARN , Animaux , Humains , Souris , Cellules 3T3-L1 , Adipocytes/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Vésicules extracellulaires/métabolisme , Régulation de l'expression des gènes , Hépatocytes/métabolisme , Ribonucléoprotéine nucléaire hétérogène A1/métabolisme , Ribonucléoprotéine nucléaire hétérogène A1/génétique , Insuline/métabolisme , Insulinorésistance , microARN/métabolisme , microARN/génétique , Obésité/métabolisme , Obésité/génétique , Phosphorylation , Transduction du signal
12.
Phytochemistry ; 226: 114206, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38972440

RÉSUMÉ

Eighteen compounds including eleven previously undescribed diterpenes were isolated from the leaves of Croton mangelong. The structures were determined by HRESIMS, IR, NMR, X-ray diffraction and ECD spectroscopic analysis. All isolates were assayed for their anti-hyperglycemic activities in insulin resistance (IR) 3T3-L1 adipocytes, and compound 4 was tested for its anti-diabetic activity in vivo. Results suggested compound 4 could effectively reduce blood glucose level in diabetic SD rats in a dose of 30 mg/kg.


Sujet(s)
Cellules 3T3-L1 , Croton , Diterpènes , Hypoglycémiants , Feuilles de plante , Rat Sprague-Dawley , Feuilles de plante/composition chimique , Diterpènes/pharmacologie , Diterpènes/composition chimique , Diterpènes/isolement et purification , Croton/composition chimique , Animaux , Souris , Hypoglycémiants/pharmacologie , Hypoglycémiants/composition chimique , Hypoglycémiants/isolement et purification , Rats , Structure moléculaire , Diabète expérimental/traitement médicamenteux , Mâle , Insulinorésistance , Glycémie/effets des médicaments et des substances chimiques , Adipocytes/effets des médicaments et des substances chimiques
13.
Adipocyte ; 13(1): 2381262, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39039652

RÉSUMÉ

Obesity is a low-grade chronic inflammation induced by the pathological expansion of adipocytes which allows the development of obesity-associated metabolic diseases like type 2 diabetes mellitus (T2D) and non-alcoholic fatty liver disease (NAFLD). However, mechanisms regulating adipocyte inflammation remain poorly understood. Here, we observed that TRIM8 was upregulated in adipocyte inflammation and insulin resistance while DUSP14 was downregulated. TRIM8 deficiency and DUSP14 over-expression decreased the level of inflammatory cytokines, increased glucose uptake content, and improved insulin signalling transduction compared to LPS treatment alone. Conversely, silencing DUSP14 increased the expression of inflammatory cytokines. It decreased the glucose uptake content and the phosphorylation level of proteins involved in insulin signalling, further impairing insulin signalling and aggravating insulin resistance. Furthermore, The decreased level of inflammatory cytokines, increased glucose uptake, and improved insulin signalling transduction caused by TRIM8 deficiency were reversed by down-regulated DUSP14. Collectively, our findings revealed that TRIM8 can regulate adipocyte inflammation and insulin resistance by regulating the MAPKs pathway which is dependent on DUSP14.


Sujet(s)
Adipocytes , Dual-specificity phosphatases , Inflammation , Insulinorésistance , Animaux , Adipocytes/métabolisme , Souris , Inflammation/métabolisme , Dual-specificity phosphatases/métabolisme , Dual-specificity phosphatases/génétique , Système de signalisation des MAP kinases , Cellules 3T3-L1 , Transduction du signal , Mitogen-Activated Protein Kinase Phosphatases/métabolisme , Mitogen-Activated Protein Kinase Phosphatases/génétique , Souris de lignée C57BL
14.
J Mater Chem B ; 12(32): 7905-7914, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39028265

RÉSUMÉ

Obesity has emerged as a significant global health burden, exacerbated by serious side effects associated with existing anti-obesity medications. Celastrol (CLT) holds promise for weight loss but encounters challenges related to poor solubility and systemic toxicity. Here, we present chondroitin sulfate (CS)-derived micelles engineered for adipocyte-specific targeting, aiming to enhance the therapeutic potential of CLT while minimizing its systemic toxicity. To further enhance adipocyte affinity, we introduced a biguanide moiety into a micellar vehicle. CS is sequentially modified with hydrophilic metformin and hydrophobic 4-aminophenylboronic acid pinacol ester (PBE), resulting in the self-assembly of CLT-encapsulated micelles (MET-CS-PBE@CLT). This innovative design imparts amphiphilicity via the PBE moieties while ensuring the outward exposure of hydrophilic metformin moieties, facilitating active interactions with adipocytes. In vitro studies confirmed the enhanced uptake of MET-CS-PBE@CLT micelles by adipocytes, while in vivo studies demonstrated increased distribution within adipose tissues. In a diet-induced obese mouse model, MET-CS-PBE@CLT exhibited remarkable efficacy in weight loss without affecting food intake. This pioneering strategy offers a promising, low-risk, and highly effective solution to address the global obesity epidemic.


Sujet(s)
Adipocytes , Micelles , Obésité , Triterpènes pentacycliques , Animaux , Triterpènes pentacycliques/composition chimique , Triterpènes pentacycliques/pharmacologie , Souris , Adipocytes/effets des médicaments et des substances chimiques , Obésité/traitement médicamenteux , Biguanides/composition chimique , Biguanides/pharmacologie , Biguanides/usage thérapeutique , Souris de lignée C57BL , Cellules 3T3-L1 , Systèmes de délivrance de médicaments , Mâle , Agents antiobésité/pharmacologie , Agents antiobésité/composition chimique , Taille de particule , Chondroïtines sulfate/composition chimique , Vecteurs de médicaments/composition chimique , Triterpènes/composition chimique , Triterpènes/pharmacologie
15.
Biochem Biophys Res Commun ; 731: 150279, 2024 Oct 30.
Article de Anglais | MEDLINE | ID: mdl-39018972

RÉSUMÉ

This study assessed the anti-obesity effects of Lactobacillus paracasei subsp. paracasei NTU 101 (NTU 101) both in vitro and in vivo. Initially, the cytotoxicity and lipid accumulation inhibitory effects of NTU 101 on 3T3-L1 cells were evaluated using the MTT assay and oil red O assay, respectively. Subsequently, the anti-obesity effects of NTU 101 were investigated in high-fat diet-induced obese rat. Moreover, western blotting was performed to measure the obesity-related protein expression of PPARα, PPARß, PPARγ, C/EBPα, C/EBPß, ATGL, p-p38 MAPK, p-ERK1/2, p-AMPK and CPT-1 in both 3T3-L1 adipocytes and adipose and liver tissues. Treatment with 16 × 108 CFU/mL NTU 101 reduced lipid accumulation in 3T3-L1 adipocytes by more than 50 %. Oral administration of NTU 101 significantly attenuated body weight gain, as well as adipose tissue weight. NTU 101 administration enhanced fatty acid oxidation increasing expression levels of PPARα, CPT-1, and p-AMPK proteins in liver tissue, while simultaneously inhibited adipogenesis by reducing PPARγ and C/EBPα proteins in adipose tissue. Furthermore, NTU 101 supplementation positively modulated the composition of gut microbiota, notably increasing the abundance of Akkermansiaceae. This present study suggests that NTU 101 exerts anti-obesity effects by regulating gut microbiota, fatty acid oxidation, lipolysis and adipogenesis.


Sujet(s)
Cellules 3T3-L1 , AMP-Activated Protein Kinases , Microbiome gastro-intestinal , Lacticaseibacillus paracasei , Obésité , Probiotiques , Animaux , Obésité/métabolisme , Obésité/microbiologie , Obésité/prévention et contrôle , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Souris , Lacticaseibacillus paracasei/métabolisme , Mâle , Rats , AMP-Activated Protein Kinases/métabolisme , Probiotiques/administration et posologie , Probiotiques/pharmacologie , Rat Sprague-Dawley , Alimentation riche en graisse/effets indésirables , Transduction du signal/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Métabolisme lipidique/effets des médicaments et des substances chimiques , Foie/métabolisme , Agents antiobésité/pharmacologie
16.
Nutr Res ; 128: 14-23, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39002358

RÉSUMÉ

Sesamolin, a lignan isolated from sesame oils, has been found to possess neuroprotective, anticancer, and free radical scavenging properties. We hypothesized that sesamolin could stimulate the activity of nuclear factor erythroid-derived 2-like 2 (Nrf2) and inhibit adipocyte differentiation of preadipocytes. The objective of this study was to investigate effects of sesamolin on adipocyte differentiation and its underlying molecular mechanisms. In this study, we determined the effects of treatment with 25 to 100 µM sesamolin on adipogenesis in cell culture systems. Sesamolin inhibited lipid accumulation and suppressed the expression of adipocyte markers during adipocyte differentiation of C3H10T1/2, 3T3-L1, and primary preadipocytes. Mechanism studies revealed that sesamolin increased Nrf2 protein expression without inducing its mRNA, leading to an increase in the expression of Nrf2 target genes such as heme oxygenase 1 and NAD(P)H:quinone oxidoreductase 1 (Nqo1) in C3H10T1/2 adipocytes and mouse embryonic fibroblasts. These effects were significantly attenuated in Nrf2 knockout (KO) mouse embryonic fibroblasts, indicating that effects of sesamolin were dependent on Nrf2. In H1299 human lung cancer cells with KO of Kelch like-ECH-associated protein 1 (Keap1), a negative regulator of Nrf2, sesamolin failed to further increase Nrf2 protein expression. However, upon reexpressing Keap1 in Keap1 KO cells, the ability of sesamolin to elevate Nrf2 protein expression was restored, highlighting the crucial role of Keap1 in sesamolin-induced Nrf2 activation. Taken together, these findings show that sesamolin can inhibit adipocyte differentiation through Keap1-mediated Nrf2 activation.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Adipogenèse , Différenciation cellulaire , Protéine-1 de type kelch associée à ECH , NADPH dehydrogenase (quinone) , Facteur-2 apparenté à NF-E2 , Facteur-2 apparenté à NF-E2/métabolisme , Protéine-1 de type kelch associée à ECH/métabolisme , Animaux , Souris , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Adipogenèse/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , NADPH dehydrogenase (quinone)/métabolisme , Dioxoles/pharmacologie , Souris knockout , Lignanes/pharmacologie , Humains , Heme oxygenase-1/métabolisme , Heme oxygenase-1/génétique , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique
17.
Biochem Pharmacol ; 227: 116452, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39059772

RÉSUMÉ

The white adipose tissue-specific aptamer Adipo8 can specificity bindwith mature adipocytes or tissues and inhibit adipogenesis.In this research, we exploredthe effect of Adipo8 intervention on the transcriptome in the process of adipogenesis using mRNA-level sequencing,analyzed the mechanism ofAdipo8 ininhibiting adipogenesis. The results showed that Adipo8 can inhibit lipid formation and downregulate PPARγ and C/EBPα in differentiated 3 T3-L1 cells. Transcriptome mRNA sequencing of 3 T3-L1 cells after Adipo8 interventionrevealed that Adipo8 might inhibit the biological function of adipogenesis by downregulating Acsl1 and Plin1 to inhibit fatty acid metabolism and PPAR signaling pathways.After that, using Spacer18 to connect the optimized and truncated Adipo8, we constructed a bivalent aptamer Adipo8cBand compared the affinity, biological effects, and biological stability between the aptamers in differentiated and mature 3 T3-L1 cells. At the cellular level,the affinity, biological effects, and serum stability of Adipo8cB were verified to be superior to those of Adipo8in 3 T3-L1 cells.We then investigated the biological properties of Adipo8cB as a lipid-inhibiting drug invivo, using C57BL/6J mice with diet-induced obesity. The body weight, blood sugar, lipid levels, liver function, glucose tolerance, and other related indicators in each group of mice were observed and compared after intervention with the bivalent aptamers Adipo8cB and Adipo8. Both Adipo8cB and Adipo8 effectively prevented weight gain caused by fat accumulation in micewith diet induced obesity, while also reducing blood lipid levels, improving glucose tolerance, and protecting against liver steatosis, moreover, Adipo8cB has a better effect than Adipo8.


Sujet(s)
Cellules 3T3-L1 , Tissu adipeux blanc , Aptamères nucléotidiques , Souris de lignée C57BL , Obésité , Animaux , Souris , Obésité/métabolisme , Tissu adipeux blanc/métabolisme , Tissu adipeux blanc/effets des médicaments et des substances chimiques , Aptamères nucléotidiques/pharmacologie , Mâle , Adipogenèse/effets des médicaments et des substances chimiques , Adipogenèse/physiologie , Alimentation riche en graisse/effets indésirables
18.
Article de Anglais | MEDLINE | ID: mdl-38844189

RÉSUMÉ

In this study, we focused on confirming the steroid hormone receptor-mediated endocrine-disrupting potential of the pyrethroid insecticide fenvalerate and unraveling the underlying mechanisms. Therefore, we assessed estrogen receptor-α (ERα)- and androgen receptor (AR)-mediated responses in vitro using a hormone response element-dependent transcription activation assay with a luciferase reporter following the Organization for Economic Cooperation and Development (OECD) test guidelines. We observed that fenvalerate acted as estrogen by inducing the translocation of cytosolic ERα to the nucleus via ERα dimerization, whereas it exhibited no AR-mediated androgen response element-dependent luciferase activity. Furthermore, we confirmed that fenvalerate-induced activation of ERα caused lipid accumulation, promoted in a fenvalerate-dependent manner in 3 T3-L1 adipocytes. Moreover, fenvalerate-induced lipid accumulation was inhibited in the presence of an ERα-selective antagonist, whereas it remained unaffected in the presence of a glucocorticoid receptor (GR)-specific inhibitor. In addition, fenvalerate was found to stimulate the expression of transcription factors that promote lipid accumulation in 3 T1-L1 adipocytes, and co-treatment with an ERα-selective antagonist suppressed adipogenic/ lipogenic transcription factors at both mRNA and protein levels. These findings suggest that fenvalerate exposure may lead to lipid accumulation by interfering with ERα activation-dependent processes, thus causing an ERα-mediated endocrine-disrupting effect.


Sujet(s)
Cellules 3T3-L1 , Perturbateurs endocriniens , Récepteur alpha des oestrogènes , Nitriles , Pyréthrines , Pyréthrines/toxicité , Animaux , Nitriles/toxicité , Souris , Perturbateurs endocriniens/toxicité , Récepteur alpha des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/génétique , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Métabolisme lipidique/effets des médicaments et des substances chimiques , Récepteurs aux androgènes/métabolisme , Insecticides/toxicité , Organisation de coopération et de développement économiques
19.
Biochem Pharmacol ; 226: 116381, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38909786

RÉSUMÉ

The escalating prevalence of obesity presents formidable challenges, necessitating the development of effective therapeutic strategies. In this study, we aimed to elucidate the preventive effects on obesity of tetrahydroberberrubine (THBru), a derivative of berberine (BBR) and to unravel its underlying mechanism. Using an obese mouse model induced by a high-fat diet (HFD), THBru was found to markedly ameliorate obesity, as evidenced by reduced body weight, decreased Lee's index, diminished fat mass in epididymal white adipose tissue (WAT) and brown adipose tissue (BAT), alongside improved dyslipidemia. Notably, at the same dose, THBru exhibited superior efficacy compared to BBR. RNA-sequencing and gene set enrichment analysis indicated THBru activated thermogenesis, which was further confirmed in WAT, BAT, and 3T3-L1 cells. Bioinformatics analysis of RNA-sequencing data revealed the candidate gene Pgc1α, a key regulator involved in thermogenesis. Moreover, THBru was demonstrated to elevate the expression of PGC1α by stabilizing its mRNA in WAT, BAT and 3T3-L1 cells. Furthermore, PGC1α knockdown blocked the pro-thermogenic and anti-obesity action of THBru both in vivo and in vitro. This study unravels the preventive effects of THBru on obesity through the activation of PGC1α-mediated thermogenesis, thereby delineating its potential therapeutic implications for obesity and associated disorders.


Sujet(s)
Cellules 3T3-L1 , Tissu adipeux brun , Tissu adipeux blanc , Berbérine , Alimentation riche en graisse , Souris de lignée C57BL , Obésité , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Thermogenèse , Animaux , Thermogenèse/effets des médicaments et des substances chimiques , Souris , Tissu adipeux brun/effets des médicaments et des substances chimiques , Tissu adipeux brun/métabolisme , Mâle , Berbérine/pharmacologie , Berbérine/analogues et dérivés , Berbérine/usage thérapeutique , Obésité/prévention et contrôle , Obésité/métabolisme , Obésité/traitement médicamenteux , Tissu adipeux blanc/effets des médicaments et des substances chimiques , Tissu adipeux blanc/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Alimentation riche en graisse/effets indésirables , Agents antiobésité/pharmacologie
20.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38928498

RÉSUMÉ

Extensive evidence supports the connection between obesity-induced inflammation and the heightened expression of IL-6 adipose tissues. However, the mechanism underlying the IL-6 exacerbation in the adipose tissue remains unclear. There is general agreement that TNF-α and stearate concentrations are mildly elevated in adipose tissue in the state of obesity. We hypothesize that TNF-α and stearate co-treatment induce the increased expression of IL-6 in mouse adipocytes. We therefore aimed to determine IL-6 gene expression and protein production by TNF-α/stearate treated adipocytes and investigated the mechanism involved. To test our hypothesis, 3T3-L1 mouse preadipocytes were treated with TNF-α, stearate, or TNF-α/stearate. IL-6 gene expression was assessed by quantitative real-time qPCR. IL-6 protein production secreted in the cell culture media was determined by ELISA. Acetylation of histone was analyzed by Western blotting. Il6 region-associated histone H3 lysine 9/18 acetylation (H3K9/18Ac) was determined by ChIP-qPCR. 3T3-L1 mouse preadipocytes were co-challenged with TNF-α and stearate for 24 h, which led to significantly increased IL-6 gene expression (81 ± 2.1 Fold) compared to controls stimulated with either TNF-α (38 ± 0.5 Fold; p = 0.002) or stearate (56 ± 2.0 Fold; p = 0.013). As expected, co-treatment of adipocytes with TNF-α and stearate significantly increased protein production (338 ± 11 pg/mL) compared to controls stimulated with either TNF-α (28 ± 0.60 pg/mL; p = 0.001) or stearate (53 ± 0.20 pg/mL, p = 0.0015). Inhibition of histone acetyltransferases (HATs) with anacardic acid or curcumin significantly reduced the IL-6 gene expression and protein production by adipocytes. Conversely, TSA-induced acetylation substituted the stimulatory effect of TNF-α or stearate in their synergistic interaction for driving IL-6 gene expression and protein production. Mechanistically, TNF-α/stearate co-stimulation increased the promoter-associated histone H3 lysine 9/18 acetylation (H3K9/18Ac), rendering a transcriptionally permissive state that favored IL-6 expression at the transcriptional and translational levels. Our data represent a TNF-α/stearate cooperativity model driving IL-6 expression in 3T3-L1 cells via the H3K9/18Ac-dependent mechanism, with implications for adipose IL-6 exacerbations in obesity.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Histone , Interleukine-6 , Facteur de nécrose tumorale alpha , Animaux , Souris , Acétylation , Adipocytes/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Histone/métabolisme , Interleukine-6/métabolisme , Interleukine-6/génétique , Acides stéariques/pharmacologie , Acides stéariques/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE