Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.676
Filtrer
1.
Am J Physiol Endocrinol Metab ; 327(1): E69-E80, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38717361

RÉSUMÉ

Acylglycerophosphate acyltransferases (AGPATs) catalyze the de novo formation of phosphatidic acid to synthesize glycerophospholipids and triglycerides. AGPATs demonstrate unique physiological roles despite a similar biochemical function. AGPAT3 is highly expressed in the testis, kidney, and liver, with intermediate expression in adipose tissue. Loss of AGPAT3 is associated with reproductive abnormalities and visual dysfunction. However, the role of AGPAT3 in adipose tissue and whole body metabolism has not been investigated. We found that male Agpat3 knockout (KO) mice exhibited reduced body weights with decreased white and brown adipose tissue mass. Such changes were less pronounced in the female Agpat3-KO mice. Agpat3-KO mice have reduced plasma insulin growth factor 1 (IGF1) and insulin levels and diminished circulating lipid metabolites. They manifested intact glucose homeostasis and insulin sensitivity despite a lean phenotype. Agpat3-KO mice maintained an energy balance with normal food intake, energy expenditure, and physical activity, except for increased water intake. Their adaptive thermogenesis was also normal despite reduced brown adipose mass and triglyceride content. Mechanistically, Agpat3 was elevated during mouse and human adipogenesis and enriched in adipocytes. Agpat3-knockdown 3T3-L1 cells and Agpat3-deficient mouse embryonic fibroblasts (MEFs) have impaired adipogenesis in vitro. Interestingly, pioglitazone treatment rescued the adipogenic deficiency in Agpat3-deficient cells. We conclude that AGPAT3 regulates adipogenesis and adipose development. It is possible that adipogenic impairment in Agpat3-deficient cells potentially leads to reduced adipose mass. Findings from this work support the unique role of AGPAT3 in adipose tissue.NEW & NOTEWORTHY AGPAT3 deficiency results in male-specific growth retardation. It reduces adipose tissue mass but does not significantly impact glucose homeostasis or energy balance, except for influencing water intake in mice. Like AGPAT2, AGPAT3 is upregulated during adipogenesis, potentially by peroxisome proliferator-activated receptor gamma (PPARγ). Loss of AGPAT3 impairs adipocyte differentiation, which could be rescued by pioglitazone. Overall, AGPAT3 plays a significant role in regulating adipose tissue mass, partially involving its influence on adipocyte differentiation.


Sujet(s)
1-Acylglycerol-3-phosphate O-acyltransferase , Adipocytes , Souris knockout , Animaux , Femelle , Mâle , Souris , 1-Acylglycerol-3-phosphate O-acyltransferase/génétique , 1-Acylglycerol-3-phosphate O-acyltransferase/métabolisme , Adipocytes/métabolisme , Adipogenèse/génétique , Adipogenèse/physiologie , Tissu adipeux brun/métabolisme , Différenciation cellulaire , Métabolisme énergétique/génétique , Insulinorésistance/génétique , Souris de lignée C57BL , Phénotype , Thermogenèse/génétique , Maigreur/métabolisme , Maigreur/génétique
2.
J Endocrinol ; 262(1)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38692289

RÉSUMÉ

CD44, a cell surface adhesion receptor and stem cell biomarker, is recently implicated in chronic metabolic diseases. Ablation of CD44 ameliorates adipose tissue inflammation and insulin resistance in obesity. Here, we investigated cell type-specific CD44 expression in human and mouse adipose tissue and further studied how CD44 in preadipocytes regulates adipocyte function. Using Crispr Cas9-mdediated gene deletion and lentivirus-mediated gene re-expression, we discovered that deletion of CD44 promotes adipocyte differentiation and adipogenesis, whereas re-expression of CD44 abolishes this effect and decreases insulin responsiveness and adiponectin secretion in 3T3-L1 cells. Mechanistically, CD44 does so via suppressing Pparg expression. Using quantitative proteomics analysis, we further discovered that cell cycle-regulated pathways were mostly decreased by deletion of CD44. Indeed, re-expression of CD44 moderately restored expression of proteins involved in all phases of the cell cycle. These data were further supported by increased preadipocyte proliferation rates in CD44-deficient cells and re-expression of CD44 diminished this effect. Our data suggest that CD44 plays a crucial role in regulating adipogenesis and adipocyte function possibly through regulating PPARγ and cell cycle-related pathways. This study provides evidence for the first time that CD44 expressed in preadipocytes plays key roles in regulating adipocyte function outside immune cells where CD44 is primarily expressed. Therefore, targeting CD44 in (pre)adipocytes may provide therapeutic potential to treat obesity-associated metabolic complications.


Sujet(s)
Cellules 3T3-L1 , Adipocytes , Adipogenèse , Cycle cellulaire , Antigènes CD44 , Récepteur PPAR gamma , Adipogenèse/génétique , Adipogenèse/physiologie , Antigènes CD44/métabolisme , Antigènes CD44/génétique , Animaux , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Souris , Cycle cellulaire/génétique , Cycle cellulaire/physiologie , Humains , Adipocytes/métabolisme , Délétion de gène , Différenciation cellulaire/génétique , Mâle , Tissu adipeux/métabolisme , Tissu adipeux/cytologie , Transduction du signal/physiologie
3.
Am J Physiol Endocrinol Metab ; 327(1): E13-E26, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38717362

RÉSUMÉ

Adipose tissue metabolism is actively involved in the regulation of energy balance. Adipose-derived stem cells (ASCs) play a critical role in maintaining adipose tissue function through their differentiation into mature adipocytes (Ad). This study aimed to investigate the impact of an obesogenic environment on the epigenetic landscape of ASCs and its impact on adipocyte differentiation and its metabolic consequences. Our results showed that ASCs from rats on a high-fat sucrose (HFS) diet displayed reduced adipogenic capacity, increased fat accumulation, and formed larger adipocytes than the control (C) group. Mitochondrial analysis revealed heightened activity in undifferentiated ASC-HFS but decreased respiratory and glycolytic capacity in mature adipocytes. The HFS diet significantly altered the H3K4me3 profile in ASCs on genes related to adipogenesis, mitochondrial function, inflammation, and immunomodulation. After differentiation, adipocytes retained H3K4me3 alterations, confirming the upregulation of genes associated with inflammatory and immunomodulatory pathways. RNA-seq confirmed the upregulation of genes associated with inflammatory and immunomodulatory pathways in adipocytes. Overall, the HFS diet induced significant epigenetic and transcriptomic changes in ASCs, impairing differentiation and causing dysfunctional adipocyte formation.NEW & NOTEWORTHY Obesity is associated with the development of chronic diseases like metabolic syndrome and type 2 diabetes, and adipose tissue plays a crucial role. In a rat model, our study reveals how an obesogenic environment primes adipocyte precursor cells, leading to epigenetic changes that affect inflammation, adipogenesis, and mitochondrial activity after differentiation. We highlight the importance of histone modifications, especially the trimethylation of histone H3 to lysine 4 (H3K4me3), showing its influence on adipocyte expression profiles.


Sujet(s)
Adipocytes , Adipogenèse , Tissu adipeux , Alimentation riche en graisse , Épigenèse génétique , Histone , Transcriptome , Animaux , Rats , Adipocytes/métabolisme , Alimentation riche en graisse/effets indésirables , Histone/métabolisme , Mâle , Adipogenèse/génétique , Adipogenèse/physiologie , Tissu adipeux/métabolisme , Différenciation cellulaire/génétique , Cellules souches/métabolisme , Obésité/métabolisme , Obésité/génétique , Reprogrammation cellulaire/physiologie , Cellules cultivées , Rat Wistar , Rat Sprague-Dawley
4.
Dev Cell ; 59(10): 1231-1232, 2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38772342

RÉSUMÉ

Brown adipocytes are found in several fat depots, however, the origins and contributions of different lineages of adipogenic progenitor cells (APCs) to these depots are unclear. In this issue of Developmental Cell, Shi et al. show that platelet-derived growth factor receptor ß (PDGFRß)-lineage and T-box transcription factor 18 (TBX18)-lineage APCs differentially contribute to brown adipogenesis across these depots.


Sujet(s)
Adipogenèse , Récepteurs Notch , Cellules souches , Adipogenèse/physiologie , Animaux , Récepteurs Notch/métabolisme , Cellules souches/métabolisme , Cellules souches/cytologie , Tissu adipeux brun/métabolisme , Tissu adipeux brun/cytologie , Humains , Adipocytes bruns/métabolisme , Adipocytes bruns/cytologie , Récepteur au PDGF bêta/métabolisme , Récepteur au PDGF bêta/génétique , Différenciation cellulaire , Lignage cellulaire , Souris , Transduction du signal
5.
Pediatr Obes ; 19(6): e13120, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38590200

RÉSUMÉ

Maternal obesity is a well-known risk factor for developing premature obesity, metabolic syndrome, cardiovascular disease and type 2 diabetes in the progeny. The development of white adipose tissue is a dynamic process that starts during prenatal life: fat depots laid down in utero are associated with the proportion of fat in children later on. How early this programming takes place is still unknown. However, recent evidence shows that mesenchymal stem cells (MSC), the embryonic adipocyte precursor cells, show signatures of the early setting of an adipogenic committed phenotype when exposed to maternal obesity. This review aims to present current findings on the cellular adaptations of MSCs from the offspring of women with obesity and how the metabolic environment of MSCs could affect the early commitment towards adipocytes. In conclusion, maternal obesity can induce early programming of fetal adipose tissue by conditioning MSCs. These cells have higher expression of adipogenic markers, altered insulin signalling and mitochondrial performance, compared to MSCs of neonates from lean pregnancies. Fetal MSCs imprinting by maternal obesity could help explain the increased risk of childhood obesity and development of further noncommunicable diseases.


Sujet(s)
Cellules souches mésenchymateuses , Obésité maternelle , Effets différés de l'exposition prénatale à des facteurs de risque , Humains , Femelle , Grossesse , Obésité maternelle/métabolisme , Tissu adipeux , Obésité pédiatrique , Adipogenèse/physiologie , Nouveau-né , Adipocytes
6.
STAR Protoc ; 5(2): 103019, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38635394

RÉSUMÉ

In vitro cell culture serves as an efficient system for studying animal cell behavior in a controlled setting. Here, we present a 3D culture model for forming ruminant adipose organoids using stromal vascular fraction cells. We describe steps for forming cell spheroids and growing them on a Matrigel-coated surface. We then detail procedures for inducing organoids to undergo angiogenesis and adipogenesis followed by capillary sprouting. This protocol can be utilized to study the interaction between blood vessels and adipocytes. For complete details on the use and execution of this protocol, please refer to Yu et al.1.


Sujet(s)
Tissu adipeux , Organoïdes , Animaux , Organoïdes/cytologie , Tissu adipeux/cytologie , Cellules stromales/cytologie , Ruminants , Techniques de culture cellulaire/méthodes , Néovascularisation physiologique/physiologie , Adipogenèse/physiologie , Adipocytes/cytologie , Cellules cultivées
7.
Poult Sci ; 103(6): 103728, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38688194

RÉSUMÉ

E2F transcription factor 5 (E2F5) gene is a transcription factor, plays an important role in the development of a variety of cells. E2F5 is expressed in human and mouse adipocytes, but its specific function in adipogenesis is unclear. Krüppel-like factor 7 (KLF7) facilitates proliferation and inhibits differentiation in chicken preadipocytes. Our previous KLF7 chromatin immunoprecipitation-sequencing analysis revealed a KLF7-binding peak in the 3' flanking region of the E2F5, indicating a regulatory role of KLF7 in this region. In the present study, we investigated E2F5 potential role, the overexpression and knockdown analyses revealed that E2F5 inhibited the differentiation and promoted the proliferation of chicken preadipocytes. Moreover, we identified enhancer activity in the 3' flanking region (nucleotides +22661/+22900) of E2F5 and found that KLF7 overexpression increased E2F5 expression and luciferase activity in this region. Deleting the putative KLF7-binding site eliminated the promoting effect of KLF7 overexpression on E2F5 expression. Further, E2F5 reversed the KLF7-induced decrease in preadipocyte differentiation and increase in preadipocyte proliferation. Taken together, our findings demonstrate that KLF7 inhibits differentiation and promotes proliferation in preadipocytes by enhancing E2F5 transcription.


Sujet(s)
Adipocytes , Adipogenèse , Différenciation cellulaire , Prolifération cellulaire , Poulets , Facteurs de transcription Krüppel-like , Animaux , Facteurs de transcription Krüppel-like/génétique , Facteurs de transcription Krüppel-like/métabolisme , Adipogenèse/physiologie , Poulets/génétique , Adipocytes/métabolisme , Adipocytes/physiologie , Facteur de transcription E2F5/métabolisme , Facteur de transcription E2F5/génétique , Facteur de transcription E2F5/physiologie , Protéines aviaires/métabolisme , Protéines aviaires/génétique
8.
Am J Physiol Cell Physiol ; 326(6): C1611-C1624, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38646789

RÉSUMÉ

The influence of SGLT-1 on perivascular preadipocytes (PVPACs) and vascular remodeling is not well understood. This study aimed to elucidate the role and mechanism of SGLT-1-mediated PVPACs bioactivity. PVPACs were cultured in vitro and applied ex vivo to the carotid arteries of mice using a lentivirus-based thermosensitive in situ gel (TISG). The groups were treated with Lv-SGLT1 (lentiviral vector, overexpression), Lv-siSGLT1 (RNA interference, knockdown), or specific signaling pathway inhibitors. Assays were conducted to assess changes in cell proliferation, apoptosis, glucose uptake, adipogenic differentiation, and vascular remodeling in the PVPACs. Protein expression was analyzed by Western blotting, immunocytochemistry, and/or immunohistochemistry. The methyl thiazolyl tetrazolium (MTT) assay and Hoechst 33342 staining indicated that SGLT-1 overexpression significantly promoted PVPACs proliferation and inhibited apoptosis in vitro. Conversely, SGLT-1 knockdown exerted the opposite effect. Oil Red O staining revealed that SGLT-1 overexpression facilitated adipogenic differentiation, while its inhibition mitigated these effects. 3H-labeled glucose uptake experiments demonstrated that SGLT-1 overexpression enhanced glucose uptake by PVPACs, whereas RNA interference-mediated SGLT-1 inhibition had no significant effect on glucose uptake. Moreover, RT-qPCR, Western blotting, and immunofluorescence analyses revealed that SGLT-1 overexpression upregulated FABP4 and VEGF-A levels and activated the Akt/mTOR/p70S6K signaling pathway, whereas SGLT-1 knockdown produced the opposite effects. In vivo studies corroborated these findings and indicated that SGLT-1 overexpression facilitated carotid artery remodeling. Our study demonstrates that SGLT-1 activation of the Akt/mTOR/p70S6K signaling pathway promotes PVPACs proliferation, adipogenesis, glucose uptake, glucolipid metabolism, and vascular remodeling.NEW & NOTEWORTHY SGLT-1 is expressed in PVPACs and can affect preadipocyte glucolipid metabolism and vascular remodeling. SGLT-1 promotes the biofunctions of PVPACs mediated by Akt/mTOR/p70S6K signaling pathway. Compared with caudal vein or intraperitoneal injection, the external application of lentivirus-based thermal gel around the carotid artery is an innovative attempt at vascular remodeling model, it may effectively avoid the transfection of lentiviral vector into the whole body of mice and the adverse effect on experimental results.


Sujet(s)
Adipocytes , Prolifération cellulaire , Protéines proto-oncogènes c-akt , Ribosomal Protein S6 Kinases, 70-kDa , Transduction du signal , Transporteur-1 sodium-glucose , Sérine-thréonine kinases TOR , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Souris , Ribosomal Protein S6 Kinases, 70-kDa/métabolisme , Ribosomal Protein S6 Kinases, 70-kDa/génétique , Adipocytes/métabolisme , Transporteur-1 sodium-glucose/métabolisme , Transporteur-1 sodium-glucose/génétique , Mâle , Adipogenèse/physiologie , Souris de lignée C57BL , Remodelage vasculaire , Cellules cultivées , Apoptose , Différenciation cellulaire , Glucose/métabolisme , Glucose/déficit
9.
Sci Data ; 11(1): 290, 2024 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-38472209

RÉSUMÉ

Fat infiltration in skeletal muscle is now recognized as a standard feature of aging and is directly related to the decline in muscle function. However, there is still a limited systematic integration and exploration of the mechanisms underlying the occurrence of myosteatosis in aging across species. Here, we re-analyzed bulk RNA-seq datasets to investigate the association between fat infiltration in skeletal muscle and aging. Our integrated analysis of single-nucleus transcriptomics in aged humans and Laiwu pigs with high intramuscular fat content, identified species-preference subclusters and revealed core gene programs associated with myosteatosis. Furthermore, we found that fibro/adipogenic progenitors (FAPs) had potential capacity of differentiating into PDE4D+/PDE7B+ preadipocytes across species. Additionally, cell-cell communication analysis revealed that FAPs may be associated with other adipogenic potential clusters via the COL4A2 and COL6A3 pathways. Our study elucidates the correlation mechanism between aging and fat infiltration in skeletal muscle, and these consensus signatures in both humans and pigs may contribute to increasing reproducibility and reliability in future studies involving in the field of muscle research.


Sujet(s)
Adipogenèse , Vieillissement , Muscles squelettiques , Sujet âgé , Animaux , Humains , Adipogenèse/physiologie , Différenciation cellulaire , Muscles squelettiques/physiologie , Suidae , Jeux de données comme sujet , RNA-Seq , Transcriptome , Adipocytes , Cellules souches
10.
Am J Physiol Endocrinol Metab ; 326(5): E709-E722, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38416071

RÉSUMÉ

Obesity and its related metabolic complications represent a significant global health challenge. Central to this is the dysregulation of glucolipid metabolism, with a predominant focus on glucose metabolic dysfunction in the current research, whereas adipose metabolism impairment garners less attention. Exosomes (EXs), small extracellular vesicles (EVs) secreted by various cells, have emerged as important mediators of intercellular communication and have the potential to be biomarkers, targets, and therapeutic tools for diverse diseases. In particular, EXs have been found to play a role in adipose metabolism by transporting cargoes such as noncoding RNAs (ncRNA), proteins, and other factors. This review article summarizes the current understanding of the role of EXs in mediating adipose metabolism disorders in obesity. It highlights their roles in adipogenesis (encompassing adipogenic differentiation and lipid synthesis), lipid catabolism, lipid transport, and white adipose browning. The insights provided by this review offer new avenues for developing exosome-based therapies to treat obesity and its associated comorbidities.


Sujet(s)
Adipogenèse , Tissu adipeux , Exosomes , Métabolisme lipidique , Obésité , Exosomes/métabolisme , Humains , Obésité/métabolisme , Tissu adipeux/métabolisme , Animaux , Adipogenèse/physiologie , Métabolisme lipidique/physiologie
11.
Life Sci ; 341: 122491, 2024 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-38336275

RÉSUMÉ

The World Health Organization (WHO) defines obesity as an urgency for health and a social emergency. Today around 39 % of people is overweight, of these over 13 % is obese. It is well-consolidated that the adipose cells are deputy to lipid storage under caloric excess; however, despite the classical idea that adipose tissue has exclusively a passive function, now it is known to be deeply involved in the regulation of systemic metabolism in physiological as well as under obesogenic conditions, with consequences on cardiovascular health. Beside two traditional types of adipose cells (white and brown), recently the beige one has been highlighted as the consequence of the healthy remodeling of white adipocytes, confirming their metabolic adaptability. In this direction, pharmacological, nutraceutical and nutrient-based approaches are addressed to positively influence inflammation and metabolism, thus contributing to reduce the obese-associated cardiovascular risk. In this scenario, hydrogen sulfide emerges as a new mediator that may regulate crucial targets involved in the regulation of metabolism. The current evidence demonstrates that hydrogen sulfide may induce peroxisome proliferator activated receptor γ (PPARγ), a crucial mediator of adipogenesis, inhibit the phosphorylation of perlipin-1 (plin-1), a protein implicated in the lipolysis, and finally promote browning process, through the release of irisin from skeletal muscle. The results summarized in this review suggest an important role of hydrogen sulfide in the regulation of metabolism and in the prevention/treatment of obese-associated cardiovascular diseases and propose new insight on the putative mechanisms underlying the release of hydrogen sulfide or its biosynthesis, delineating a further exciting field of application.


Sujet(s)
Sulfure d'hydrogène , Métabolisme lipidique , Humains , Sulfure d'hydrogène/métabolisme , Adipogenèse/physiologie , Adipocytes blancs/métabolisme , Obésité/métabolisme , Tissu adipeux brun/métabolisme , Tissu adipeux blanc/métabolisme
12.
Mol Metab ; 80: 101870, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38184275

RÉSUMÉ

OBJECTIVE: Bone morphogenetic protein (BMP) signaling is intricately involved in adipose tissue development. BMP7 together with BMP4 have been implicated in brown adipocyte differentiation but their roles during development remains poorly specified. Matrix Gla protein (MGP) inhibits BMP4 and BMP7 and is expressed in endothelial and progenitor cells. The objective was to determine the role of MGP in brown adipose tissue (BAT) development. METHODS: The approach included global and cell-specific Mgp gene deletion in combination with RNA analysis, immunostaining, thermogenic activity, and in vitro studies. RESULTS: The results revealed that MGP directs brown adipogenesis at two essential steps. Endothelial-derived MGP limits triggering of white adipogenic differentiation in the perivascular region, whereas MGP derived from adipose cells supports the transition of CD142-expressing progenitor cells to brown adipogenic maturity. Both steps were important to optimize the thermogenic function of BAT. Furthermore, MGP derived from both sources impacted vascular growth. Reduction of MGP in either endothelial or adipose cells expanded the endothelial cell population, suggesting that MGP is a factor in overall plasticity of adipose tissue. CONCLUSION: MGP displays a dual and cell-specific function in BAT, essentially creating a "cellular shuttle" that coordinates brown adipogenic differentiation with vascular growth during development.


Sujet(s)
Adipocytes bruns , , Adipocytes bruns/métabolisme , Différenciation cellulaire , Tissu adipeux brun/métabolisme , Adipogenèse/physiologie
13.
Genes (Basel) ; 15(1)2024 01 19.
Article de Anglais | MEDLINE | ID: mdl-38275607

RÉSUMÉ

Pericytes (PCs) are located surrounding the walls of small blood vessels, particularly capillaries and microvessels. In addition to their functions in maintaining vascular integrity, participating in angiogenesis, and regulating blood flow, PCs also serve as a reservoir for multi-potent stem/progenitor cells in white, brown, beige, and bone marrow adipose tissues. Due to the complex nature of this cell population, the identification and characterization of PCs has been challenging. A comprehensive understanding of the heterogeneity of PCs may enhance their potential as therapeutic targets for metabolic syndromes or bone-related diseases. This mini-review summarizes multiple PC markers commonly employed in lineage-tracing studies, with an emphasis on their contribution to adipogenesis and functions in different adipose depots under diverse metabolic conditions.


Sujet(s)
Adipogenèse , Péricytes , Adipogenèse/physiologie , Tissu adipeux , Cellules souches/métabolisme , Vaisseaux capillaires
14.
Life Sci ; 336: 122336, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-38092142

RÉSUMÉ

AIMS: Akr1A1 is a glycolytic enzyme catalyzing the reduction of aldehyde to alcohol. This study aims to delineate the role of Akr1A1 in regulating the adipo-osteogenic lineage differentiation of mesenchymal stem cells (MSCs). MAIN METHODS: MSCs derived from human bone marrow and Wharton Jelly together with gain- and loss-of-function analysis as well as supplementation with the S-Nitrosoglutathione reductase (GSNOR) inhibitor N6022 were used to study the function of Akr1A1 in controlling MSC lineage differentiation into osteoblasts and adipocytes. KEY FINDINGS: Akr1A1 expression, PKM2 activity, and lactate production were found to be decreased in osteoblast-committed MSCs, but PGC-1α increased to induce mitochondrial oxidative phosphorylation. Increased Akr1A1 inhibited the SIRT1-dependent pathway for decreasing the expressions of PGC-1α and TAZ but increasing PPAR γ in adipocyte-committed MSCs, hence promoting glycolysis in adipogenesis. In contrast, Akr1A1 expression, PKM2 activity and lactate production were all increased in adipocyte-differentiated cells with decreased PGC-1α for switching energy utilization to glycolytic metabolism. Reduced Akr1A1 expression in osteoblast-committed cells relieves its inhibition of SIRT1-mediated activation of PGC-1α and TAZ for facilitating osteogenesis and mitochondrial metabolism. SIGNIFICANCE: Several metabolism-involved regulators including Akr1A1, SIRT1, PPARγ, PGC-1α and TAZ were differentially expressed in osteoblast- and adipocyte-committed MSCs. More importantly, Akr1A1 was identified as a new key regulator for controlling the MSC lineage commitment in favor of adipogenesis but detrimental to osteogenesis. Such information should be useful to develop perspective new therapeutic agents to reverse the adipo-osteogenic differentiation of BMSCs, in a way to increase in osteogenesis but decrease in adipogenesis.


Sujet(s)
Adipogenèse , Cellules souches mésenchymateuses , Humains , Adipogenèse/physiologie , Ostéogenèse/physiologie , Sirtuine-1/métabolisme , Différenciation cellulaire/physiologie , Lactates/métabolisme , Aldo-keto reductases/métabolisme
15.
Biogerontology ; 25(1): 53-69, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37725294

RÉSUMÉ

With the increase of population aging, the prevalence of type 2 diabetes (T2D) is also rising. Aging affects the tissues and organs of the whole body, which is the result of various physiological and pathological processes. Adipose tissue has a high degree of plasticity and changes with aging. Aging changes the distribution of adipose tissue, affects adipogenesis, browning characteristics, inflammatory status and adipokine secretion, and increases lipotoxicity. These age-dependent changes in adipose tissue are an important cause of insulin resistance and T2D. Understanding adipose tissue changes can help promote healthy aging process. This review summarizes changes in adipose tissue ascribable to aging, with a focus on the role of aging adipose tissue in insulin resistance and T2D.


Sujet(s)
Diabète de type 2 , Insulinorésistance , Humains , Insulinorésistance/physiologie , Tissu adipeux , Vieillissement , Adipogenèse/physiologie
16.
Obes Rev ; 25(1): e13647, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37789591

RÉSUMÉ

Iron plays a vital role in essential biological processes and requires precise regulation within the body. Dysregulation of iron homeostasis, characterized by increased serum ferritin levels and excessive accumulation of iron in the liver, adipose tissue, and skeletal muscle, is associated with obesity and insulin resistance. Notably, iron excess in adipose tissue promotes adipose tissue dysfunction. As optimal adipose tissue function is crucial for maintaining a healthy phenotype in obesity, a comprehensive understanding of iron homeostasis in adipose tissue is imperative for designing new therapeutic approaches to improve and prevent adipose tissue dysfunction. Here, we conducted a review of relevant studies, focusing on and providing valuable insights into the intricate interplay between iron and adipose tissue. It sheds light on the impact of iron on adipogenesis and the physiology of both white and brown adipose tissue. Furthermore, we highlight the critical role of key modulators, such as cytosolic aconitase, mitochondria, and macrophages, in maintaining iron homeostasis within adipose tissue.


Sujet(s)
Insulinorésistance , Fer , Humains , Tissu adipeux , Tissu adipeux brun , Obésité/génétique , Adipogenèse/physiologie
17.
Int J Biochem Cell Biol ; 167: 106507, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38142772

RÉSUMÉ

Zinc finger proteins (ZFPs) constitute a crucial group of transcription factors widely present in various organisms. They act as transcription factors, nucleases, and RNA-binding proteins, playing significant roles in cell differentiation, growth, and development. With extensive research on ZFPs, their roles in the determination of mesenchymal stem cells (MSCs) fate during osteogenic and adipogenic differentiation processes have become increasingly clear. ZFP521, for instance, is identified as an inhibitor of the Wnt signaling pathway and RUNX2's transcriptional activity, effectively suppressing osteogenic differentiation. Moreover, ZFP217 contributes to the inhibition of adipogenic differentiation by reducing the M6A level of the cell cycle regulator cyclin D1 (CCND1). In addition, other ZFPs can also influence the fate of mesenchymal stem cells (MSCs) during osteogenic and adipogenic differentiation through various signaling pathways, transcription factors, and epigenetic controls, participating in the subsequent differentiation and maturation of precursor cells. Given the prevalent occurrence of osteoporosis, obesity, and related metabolic disorders, a comprehensive understanding of the regulatory mechanisms balancing bone and fat metabolism is essential, with a particular focus on the fate determination of MSCs in osteogenic and adipogenic differentiation. In this review, we provide a detailed summary of how zinc finger proteins influence the osteogenic and adipogenic differentiation of MSCs through different signaling pathways, transcription factors, and epigenetic mechanisms. Additionally, we outline the regulatory mechanisms of ZFPs in controlling osteogenic and adipogenic differentiation based on various stages of MSC differentiation.


Sujet(s)
Cellules souches mésenchymateuses , Ostéogenèse , Ostéogenèse/génétique , Différenciation cellulaire/physiologie , Adipogenèse/physiologie , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Voie de signalisation Wnt , Cellules souches mésenchymateuses/métabolisme , Doigts de zinc , Cellules cultivées
18.
Free Radic Biol Med ; 212: 65-79, 2024 02 20.
Article de Anglais | MEDLINE | ID: mdl-38141889

RÉSUMÉ

Osteoporosis is a chronic disease that seriously affects the quality of life and longevity of the elderly, so exploring the mechanism of osteoporosis is crucial for drug development and treatment. Bone marrow mesenchymal stem cells are stem cells with multiple differentiation potentials in bone marrow, and changing their differentiation direction can change bone mass. As an extracellular superoxide dismutase, Superoxide Dismutase 3 (SOD3) has been proved to play an important role in multiple organs, but the detailed mechanism of action in bone metabolism is still unclear. In this study, the results of clinical serum samples ELISA and single cell sequencing chip analysis proved that the expression of SOD3 was positively correlated with bone mass, and SOD3 was mainly expressed in osteoblasts and adipocytes and rarely expressed in osteoblasts in BMSCs. In vitro experiments showed that SOD3 can promote osteogenesis and inhibit adipogenesis. Compared with WT mice, the mice that were knocked out of SOD3 had a significant decrease in bone mineral density and significant changes in related parameters. The results of HE and IHC staining suggested that knocking out SOD3 would lead to fat accumulation in the bone marrow cavity and weakened osteogenesis. Both in vitro and in vivo experiments indicated that SOD3 affects bone metabolism by promoting osteogenesis and inhibiting adipogenesis. The results of transcriptome sequencing and revalidation showed that SOD3 can affect the expression of FLT1. Through in vitro experiments, we proved that FLT1 can also promote osteogenesis and inhibit adipogenesis. In addition, through the repeated experiments, the interaction between the two molecules (SOD3 and FLT1) was verified again. Finally, it was verified by WB that SOD3 regulates FLT1 to affect bone metabolism through PI3K/AKT and MAPK pathways.


Sujet(s)
Adipogenèse , Ostéoporose , Humains , Souris , Animaux , Sujet âgé , Adipogenèse/physiologie , Ostéogenèse/physiologie , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/génétique , Phosphatidylinositol 3-kinases/métabolisme , Qualité de vie , Différenciation cellulaire/physiologie , Ostéoporose/métabolisme , Ostéoblastes/métabolisme , Superoxide dismutase/génétique , Superoxide dismutase/métabolisme , Récepteur-1 au facteur croissance endothéliale vasculaire
19.
Int J Obes (Lond) ; 48(4): 557-566, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38148333

RÉSUMÉ

BACKGROUND: In obesity, adipose tissue undergoes a remodeling process characterized by increased adipocyte size (hypertrophia) and number (hyperplasia). The ability to tip the balance toward the hyperplastic growth, with recruitment of new fat cells through adipogenesis, seems to be critical for a healthy adipose tissue expansion, as opposed to a hypertrophic growth that is accompanied by the development of inflammation and metabolic dysfunction. However, the molecular mechanisms underlying the fine-tuned regulation of adipose tissue expansion are far from being understood. METHODS: We analyzed by mass spectrometry-based proteomics visceral white adipose tissue (vWAT) samples collected from C57BL6 mice fed with a HFD for 8 weeks. A subset of these mice, called low inflammation (Low-INFL), showed reduced adipose tissue inflammation, as opposed to those developing the expected inflammatory response (Hi-INFL). We identified the discriminants between Low-INFL and Hi-INFL vWAT samples and explored their function in Adipose-Derived human Mesenchymal Stem Cells (AD-hMSCs) differentiated to adipocytes. RESULTS: vWAT proteomics allowed us to quantify 6051 proteins. Among the candidates that most differentiate Low-INFL from Hi-INFL vWAT, we found proteins involved in adipocyte function, including adiponectin and hormone sensitive lipase, suggesting that adipocyte differentiation is enhanced in Low-INFL, as compared to Hi-INFL. The chromatin modifier SET and MYND Domain Containing 3 (SMYD3), whose function in adipose tissue was so far unknown, was another top-scored hit. SMYD3 expression was significantly higher in Low-INFL vWAT, as confirmed by western blot analysis. Using AD-hMSCs in culture, we found that SMYD3 mRNA and protein levels decrease rapidly during the adipocyte differentiation. Moreover, SMYD3 knock-down before adipocyte differentiation resulted in reduced H3K4me3 and decreased cell proliferation, thus limiting the number of cells available for adipogenesis. CONCLUSIONS: Our study describes an important role of SMYD3 as a newly discovered regulator of adipocyte precursor proliferation during the early steps of adipogenesis.


Sujet(s)
Adipocytes , Adipogenèse , Animaux , Humains , Souris , Adipocytes/métabolisme , Adipogenèse/physiologie , Tissu adipeux blanc/métabolisme , Différenciation cellulaire/génétique , Prolifération cellulaire , Histone-lysine N-methyltransferase/métabolisme , Hypertrophie/métabolisme , Inflammation/métabolisme , Souris de lignée C57BL , Obésité
20.
J Anim Sci ; 1012023 Jan 03.
Article de Anglais | MEDLINE | ID: mdl-37791975

RÉSUMÉ

Our investigation elucidated the effects of severe temperature fluctuations on cellular and physiological responses in beef cattle. Eighteen Red Angus beef steers with an average body weight of 351 ±â€…24.5 kg were divided into three treatment groups: 1) Control (CON), exposed to a temperature-humidity index (THI) of 42 for 6 h without any temperature changes; 2) Transport (TP), subjected to a one-mile trailer trip with a THI of 42 for 6 h; and 3) Temperature swing (TS), exposed to a one-mile trailer trip with a THI shift from 42 to 72-75 for 3 h. Our findings indicate that TS can induce thermal stress in cattle, regardless of whether the overall temperature level is excessively high or not. Behavioral indications of extreme heat stress in the cattle were observed, including extended tongue protrusion, reduced appetite, excessive salivation, and increased respiratory rate. Furthermore, we observed a pronounced overexpression (P < 0.05) of heat shock proteins (HSPs) 20, 27, and 90 in response to the TS treatment in the longissimus muscle (LM). Alterations in signaling pathways associated with skeletal muscle growth were noted, including the upregulation (P < 0.01) of Pax7, Myf5, and myosin heavy chain (MHC) isoforms. In addition, an increase (P < 0.05) in transcription factors associated with adipogenesis was detected (P < 0.05), such as PPARγ, C/EBPα, FAS, and SCD in the TS group, suggesting the potential for adipose tissue accumulation due to temperature fluctuations. Our data illustrated the potential impacts of these temperature fluctuations on the growth of skeletal muscle and adipose tissue in beef cattle.


In this study, we investigated the effects of severe temperature fluctuations on beef cattle and their cellular and physiological responses. Our findings demonstrate that even moderate temperature swings can cause thermal stress in cattle, leading to observable behavioral signs such as extended tongue protrusion, reduced appetite, excessive salivation, and increased respiratory rate. We also observed a significant increase in the expression of heat shock proteins (HSPs), which protect cells from stress, indicating their importance as early responders to temperature fluctuations. Furthermore, we examined the signaling pathways involved in skeletal muscle growth and found that severe temperature fluctuations can stimulate the upregulation of myogenic regulatory factors and myosin heavy chains. These changes suggest an increased demand for muscle contractile properties and hyperplasia during temperature challenges. In addition, our study revealed alterations in transcription factors associated with adipogenesis, such as PPARγ and C/EBPα, indicating the potential for adipose tissue accumulation in response to temperature fluctuations.


Sujet(s)
Adipogenèse , Tissu adipeux , Bovins , Animaux , Température , Tissu adipeux/métabolisme , Poids , Adipogenèse/physiologie , Muscles squelettiques/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...