Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.222
Filtrar
1.
Elife ; 122024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38775132

RESUMO

The energy-burning capability of beige adipose tissue is a potential therapeutic tool for reducing obesity and metabolic disease, but this capacity is decreased by aging. Here, we evaluate the impact of aging on the profile and activity of adipocyte stem and progenitor cells (ASPCs) and adipocytes during the beiging process in mice. We found that aging increases the expression of Cd9 and other fibro-inflammatory genes in fibroblastic ASPCs and blocks their differentiation into beige adipocytes. Fibroblastic ASPC populations from young and aged mice were equally competent for beige differentiation in vitro, suggesting that environmental factors suppress adipogenesis in vivo. Examination of adipocytes by single nucleus RNA-sequencing identified compositional and transcriptional differences in adipocyte populations with aging and cold exposure. Notably, cold exposure induced an adipocyte population expressing high levels of de novo lipogenesis (DNL) genes, and this response was severely blunted in aged animals. We further identified Npr3, which encodes the natriuretic peptide clearance receptor, as a marker gene for a subset of white adipocytes and an aging-upregulated gene in adipocytes. In summary, this study indicates that aging blocks beige adipogenesis and dysregulates adipocyte responses to cold exposure and provides a resource for identifying cold and aging-regulated pathways in adipose tissue.


Assuntos
Adipócitos Bege , Adipogenia , Envelhecimento , Temperatura Baixa , Animais , Adipogenia/genética , Envelhecimento/metabolismo , Envelhecimento/fisiologia , Camundongos , Adipócitos Bege/metabolismo , Camundongos Endogâmicos C57BL , Masculino , Adipócitos/metabolismo , Diferenciação Celular , Reprogramação Celular , Reprogramação Metabólica
2.
FASEB J ; 38(10): e23664, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38775797

RESUMO

Adipogenesis, a pivotal cellular process involving the differentiation of mesenchymal stem cells (MSCs) to mature adipocytes, plays a significant role in various physiological functions. Dysregulation of adipogenesis is implicated in conditions such as obesity. However, the complete molecular understanding of adipogenesis remains elusive. This study aimed to uncover the novel role of lamina-associated polypeptide 2 alpha (LAP2α) in human adipose-derived stem cells (hASCs) adipogenesis and its impact on high-fat diet (HFD)-induced obesity and associated metabolic disturbances. LAP2α expression was assessed during the adipogenic differentiation of hASCs using RT-qPCR and western blotting. The functional role of LAP2α in adipogenesis was explored both in vitro and in vivo through loss- and gain-of-function studies. Moreover, mice with HFD-induced obesity received lentivirus injection to assess the effect of LAP2α knockdown on fat accumulation. Molecular mechanisms underlying LAP2α in adipogenic differentiation were investigated using RT-qPCR, Western blotting, immunofluorescence staining, and Oil Red O staining. LAP2α expression was upregulated during hASCs adipogenic differentiation. LAP2α knockdown hindered adipogenesis, while LAP2α overexpression promoted adipogenic differentiation. Notably, LAP2α deficiency resisted HFD-induced obesity, improved glucose intolerance, mitigated insulin resistance, and prevented fatty liver development. Mechanistically, LAP2α knockdown attenuated signal transducer and activator of transcription 3 (STAT3) activation by reducing the protein level of phosphorylated STAT3. A STAT3 activator (Colivelin) counteracted the negative impact of LAP2α deficiency on hASCs adipogenic differentiation. Taken together, our current study established LAP2α as a crucial regulator of hASCs adipogenic differentiation, unveiling a new therapeutic target for obesity prevention.


Assuntos
Adipogenia , Dieta Hiperlipídica , Células-Tronco Mesenquimais , Obesidade , Humanos , Dieta Hiperlipídica/efeitos adversos , Obesidade/metabolismo , Obesidade/genética , Obesidade/etiologia , Animais , Camundongos , Células-Tronco Mesenquimais/metabolismo , Masculino , Diferenciação Celular , Camundongos Endogâmicos C57BL , Tecido Adiposo/metabolismo , Tecido Adiposo/citologia , Adipócitos/metabolismo , Células Cultivadas , Técnicas de Silenciamento de Genes , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/genética , Proteínas de Ligação a DNA , Proteínas de Membrana
3.
J Diabetes Res ; 2024: 5511454, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38736904

RESUMO

Adipose tissue dysfunction is seen among obese and type 2 diabetic individuals. Adipocyte proliferation and hypertrophy are the root causes of adipose tissue expansion. Solute carrier family 25 member 28 (SLC25A28) is an iron transporter in the inner mitochondrial membrane. This study is aimed at validating the involvement of SLC25A28 in adipose accumulation by tail vein injection of adenovirus (Ad)-SLC25A28 and Ad-green fluorescent protein viral particles into C57BL/6J mice. After 16 weeks, the body weight of the mice was measured. Subsequently, morphological analysis was performed to establish a high-fat diet (HFD)-induced model. SLC25A28 overexpression accelerated lipid accumulation in white and brown adipose tissue (BAT), enhanced body weight, reduced serum triglyceride (TG), and impaired serum glucose tolerance. The protein expression level of lipogenesis, lipolysis, and serum adipose secretion hormone was evaluated by western blotting. The results showed that adipose TG lipase (ATGL) protein expression was reduced significantly in white and BAT after overexpression SLC25A28 compared to the control group. Moreover, SLC25A28 overexpression inhibited the BAT formation by downregulating UCP-1 and the mitochondrial biosynthesis marker PGC-1α. Serum adiponectin protein expression was unregulated, which was consistent with the expression in inguinal white adipose tissue (iWAT). Remarkably, serum fibroblast growth factor (FGF21) protein expression was negatively related to the expansion of adipose tissue after administrated by Ad-SLC25A28. Data from the current study indicate that SLC25A28 overexpression promotes diet-induced obesity and accelerates lipid accumulation by regulating hormone secretion and inhibiting lipolysis in adipose tissue.


Assuntos
Adipogenia , Tecido Adiposo Marrom , Tecido Adiposo Branco , Dieta Hiperlipídica , Lipase , Camundongos Endogâmicos C57BL , Animais , Camundongos , Masculino , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Lipase/metabolismo , Lipase/genética , Obesidade/metabolismo , Lipólise , Proteína Desacopladora 1/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Transporte de Cátions/genética , Adipócitos/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Lipogênese , Aciltransferases
4.
Int J Mol Sci ; 25(9)2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38732183

RESUMO

The impact of microplastics (MPs) on the metabolic functions of the liver is currently unclear and not completely understood. To investigate the effects of the administration of MPs on the hepatic metabolism of normal and obese mice, alterations in the lipid, glucose (Glu), and amino acid regulation pathways were analyzed in the liver and adipose tissues of C57BL/6Korl (wild type, WT) or C57BL/6-Lepem1hwl/Korl mice (leptin knockout, Lep KO) orally administered polystyrene (PS) MPs for 9 weeks. Significant alterations in the lipid accumulation, adipogenesis, lipogenesis, and lipolysis pathways were detected in the liver tissue of MP-treated WT and Lep KO mice compared to the vehicle-treated group. These alterations in their liver tissues were accompanied by an upregulation of the serum lipid profile, as well as alterations in the adipogenesis, lipogenesis, and lipolysis pathways in the adipose tissues of MP-treated WT and Lep KO mice. Specifically, the level of leptin was increased in the adipose tissues of MP-treated WT mice without any change in their food intake. Also, MP-induced disruptions in the glycogenolysis, Glu transporter type 4 (GLUT4)-5' AMP-activated protein kinase (AMPK) signaling pathway, levels of lipid intermediates, and the insulin resistance of the liver tissues of WT and Lep KO mice were observed. Furthermore, the levels of seven endogenous metabolites were remarkably changed in the serum of WT and Lep KO mice after MP administrations. Finally, the impact of the MP administration observed in both types of mice was further verified in differentiated 3T3-L1 adipocytes and HepG2 cells. Thus, these results suggest that the oral administration of MPs for 9 weeks may be associated with the disruption of lipid, Glu, and amino acid metabolism in the liver tissue of obese WT and Lep KO mice.


Assuntos
Aminoácidos , Glucose , Metabolismo dos Lipídeos , Fígado , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microplásticos , Poliestirenos , Animais , Fígado/metabolismo , Fígado/efeitos dos fármacos , Camundongos , Glucose/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Aminoácidos/metabolismo , Administração Oral , Leptina/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Masculino , Lipogênese/efeitos dos fármacos , Obesidade/metabolismo , Obesidade/etiologia , Obesidade/genética , Humanos , Lipólise/efeitos dos fármacos
5.
Nutrients ; 16(9)2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38732509

RESUMO

Isoeugenol (IEG), a natural component of clove oil, possesses antioxidant, anti-inflammatory, and antibacterial properties. However, the effects of IEG on adipogenesis have not yet been elucidated. Here, we showed that IEG blocks adipogenesis in 3T3-L1 cells at an early stage. IEG inhibits lipid accumulation in adipocytes in a concentration-dependent manner and reduces the expression of mature adipocyte-related factors including PPARγ, C/EBPα, and FABP4. IEG treatment at different stages of adipogenesis showed that IEG inhibited adipocyte differentiation by suppressing the early stage, as confirmed by lipid accumulation and adipocyte-related biomarkers. The early stage stimulates growth-arrested preadipocytes to enter mitotic clonal expansion (MCE) and initiates their differentiation into adipocytes by regulating cell cycle-related factors. IEG arrested 3T3-L1 preadipocytes in the G0/G1 phase of the cell cycle and attenuated cell cycle-related factors including cyclinD1, CDK6, CDK2, and cyclinB1 during the MCE stage. Furthermore, IEG suppresses reactive oxygen species (ROS) production during MCE and inhibits ROS-related antioxidant enzymes, including superoxide dismutase1 (SOD1) and catalase. The expression of cell proliferation-related biomarkers, including pAKT and pERK1/2, was attenuated by the IEG treatment of 3T3-L1 preadipocytes. These findings suggest that it is a potential therapeutic agent for the treatment of obesity.


Assuntos
Células 3T3-L1 , Adipócitos , Adipogenia , Eugenol , Mitose , Espécies Reativas de Oxigênio , Animais , Adipogenia/efeitos dos fármacos , Camundongos , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Mitose/efeitos dos fármacos , Eugenol/farmacologia , Eugenol/análogos & derivados , Espécies Reativas de Oxigênio/metabolismo , Diferenciação Celular/efeitos dos fármacos , PPAR gama/metabolismo , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a Ácido Graxo/metabolismo , Proteínas de Ligação a Ácido Graxo/genética , Metabolismo dos Lipídeos/efeitos dos fármacos , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Antioxidantes/farmacologia
6.
Nutrients ; 16(9)2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38732619

RESUMO

Functional foods with probiotics are safe and effective dietary supplements to improve overweight and obesity. Thus, altering the intestinal microflora may be an effective approach for controlling or preventing obesity. This review aims to summarize the experimental method used to study probiotics and obesity, and recent advances in probiotics against obesity. In particular, we focused on studies (in vitro and in vivo) that used probiotics to treat obesity and its associated comorbidities. Several in vitro and in vivo (animal and human clinical) studies conducted with different bacterial species/strains have reported that probiotics promote anti-obesity effects by suppressing the differentiation of pre-adipocytes through immune cell activation, maintaining the Th1/Th2 cytokine balance, altering the intestinal microbiota composition, reducing the lipid profile, and regulating energy metabolism. Most studies on probiotics and obesity have shown that probiotics are responsible for a notable reduction in weight gain and body mass index. It also increases the levels of anti-inflammatory adipokines and decreases those of pro-inflammatory adipokines in the blood, which are responsible for the regulation of glucose and fatty acid breakdown. Furthermore, probiotics effectively increase insulin sensitivity and decrease systemic inflammation. Taken together, the intestinal microbiota profile found in overweight individuals can be modified by probiotic supplementation which can create a promising environment for weight loss along enhancing levels of adiponectin and decreasing leptin, tumor necrosis factor (TNF)-α, interleukin (IL)-6, monocyte chemotactic protein (MCP)-1, and transforming growth factor (TGF)-ß on human health.


Assuntos
Adipogenia , Anti-Inflamatórios , Microbioma Gastrointestinal , Obesidade , Probióticos , Probióticos/farmacologia , Probióticos/uso terapêutico , Humanos , Obesidade/microbiologia , Animais , Anti-Inflamatórios/farmacologia , Inflamação , Adipocinas/sangue
7.
Life Sci Alliance ; 7(7)2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38702075

RESUMO

Excess abdominal fat is a sexually dimorphic risk factor for cardio-metabolic disease and is approximated by the waist-to-hip ratio adjusted for body mass index (WHRadjBMI). Whereas this trait is highly heritable, few causal genes are known. We aimed to identify novel drivers of WHRadjBMI using systems genetics. We used two independent cohorts of adipose tissue gene expression and constructed sex- and depot-specific Bayesian networks to model gene-gene interactions from 8,492 genes. Using key driver analysis, we identified genes that, in silico and putatively in vitro, regulate many others. 51-119 key drivers in each network were replicated in both cohorts. In other cell types, 23 of these genes are found in crucial adipocyte pathways: Wnt signaling or mitochondrial function. We overexpressed or down-regulated seven key driver genes in human subcutaneous pre-adipocytes. Key driver genes ANAPC2 and RSPO1 inhibited adipogenesis, whereas PSME3 increased adipogenesis. RSPO1 increased Wnt signaling activity. In differentiated adipocytes, MIGA1 and UBR1 down-regulation led to mitochondrial dysfunction. These five genes regulate adipocyte function, and we hypothesize that they regulate fat distribution.


Assuntos
Adipócitos , Adipogenia , Distribuição da Gordura Corporal , Humanos , Adipócitos/metabolismo , Masculino , Feminino , Adipogenia/genética , Índice de Massa Corporal , Adulto , Redes Reguladoras de Genes , Pessoa de Meia-Idade , Teorema de Bayes , Relação Cintura-Quadril , Tecido Adiposo/metabolismo , Via de Sinalização Wnt/genética , Regulação da Expressão Gênica/genética , Biologia de Sistemas/métodos
8.
Anim Sci J ; 95(1): e13951, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38703069

RESUMO

Intramuscular fat (IMF) is a crucial determinant of meat quality and is influenced by various regulatory factors. Despite the growing recognition of the important role of long noncoding RNAs (lncRNAs) in IMF deposition, the mechanisms underlying buffalo IMF deposition remain poorly understood. In this study, we identified and characterized a lncRNA, lncFABP4, which is transcribed from the antisense strand of fatty acid-binding protein 4 (FABP4). lncFABP4 inhibited cell proliferation in buffalo intramuscular preadipocytes. Moreover, lncFABP4 significantly increased intramuscular preadipocyte differentiation, as indicated by an increase in the expression of the adipogenic markers peroxisome proliferator-activated receptor gamma (PPARG), CCAAT enhancer binding protein alpha (C/EBPα), and FABP4. Mechanistically, lncFABP4 was found to have the potential to regulate downstream gene expression by participating in protein-protein interaction pathways. These findings contribute to further understanding of the intricate mechanisms through which lncRNAs modulate intramuscular adipogenesis in buffaloes.


Assuntos
Adipócitos , Adipogenia , Búfalos , Diferenciação Celular , Proliferação de Células , Proteínas de Ligação a Ácido Graxo , PPAR gama , RNA Longo não Codificante , Animais , Búfalos/genética , Búfalos/metabolismo , Adipogenia/genética , Adipócitos/metabolismo , Adipócitos/citologia , Proteínas de Ligação a Ácido Graxo/metabolismo , Proteínas de Ligação a Ácido Graxo/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Diferenciação Celular/genética , PPAR gama/metabolismo , PPAR gama/genética , Expressão Gênica , Células Cultivadas , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Qualidade dos Alimentos
9.
Biochem Biophys Res Commun ; 716: 149998, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38692012

RESUMO

The equilibrium between the hypertrophic growth of existing adipocytes and adipogenesis is vital in managing metabolic stability in white adipocytes when faced with overnutrition. Adipogenesis has been established as a key player in combating metabolic irregularities caused by various factors. However, the benefits of increasing adipogenesis-mediated white adipose tissue (WAT) expansion for metabolic health regulation remain uncertain. Our findings reveal an increase in Impdh2 expression during the adipogenesis phase, both in vivo and in vitro. Xmp enhances adipogenic potential by fostering mitotic clonal expansion (MCE). The conditional knockout of Impdh2 in adipocyte progenitor cells(APCs) in adult and aged mice effectively curbs white adipose tissue expansion, ameliorates glucose tolerance, and augments energy expenditure under high-fat diet (HFD). However, no significant difference is observed under normal chow diet (NCD). Concurrently, the knockout of Impdh2 in APCs significantly reduces the count of new adipocytes induced by HFD, without affecting adipocyte size. Mechanistically, Impdh2 regulates the proliferation of APCs during the MCE phase via Xmp. Exogenous Xmp can significantly offset the reduction in adipogenic abilities of APCs due to Impdh2 deficiency. In summary, we discovered that adipogenesis-mediated WAT expansion, induced by overnutrition, also contributes to metabolic abnormalities. Moreover, the pivotal role of Impdh2 in regulating adipogenesis in APCs offers a novel therapeutic approach to combat obesity.


Assuntos
Adipócitos , Adipogenia , Tecido Adiposo Branco , Dieta Hiperlipídica , Camundongos Knockout , Hipernutrição , Animais , Tecido Adiposo Branco/metabolismo , Adipogenia/genética , Hipernutrição/metabolismo , Hipernutrição/genética , Camundongos , Adipócitos/metabolismo , Camundongos Endogâmicos C57BL , Masculino , Metabolismo Energético/genética , Deleção de Genes , Proliferação de Células , Células-Tronco/metabolismo , Células-Tronco/citologia , Células-Tronco/patologia
10.
PLoS One ; 19(5): e0298827, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38722949

RESUMO

Glutathione peroxidase 2 (GPX2) is a selenium-dependent enzyme and protects cells against oxidative damage. Recently, GPX2 has been identified as a candidate gene for backfat and feed efficiency in pigs. However, it is unclear whether GPX2 regulates the development of porcine preadipocytes and skeletal muscle cells. In this study, adenoviral gene transfer was used to overexpress GPX2. Our findings suggest that overexpression of GPX2 gene inhibited proliferation of porcine preadipocytes. And the process is accompanied by the reduction of the p-p38. GPX2 inhibited adipogenic differentiation and promoted lipid degradation, while ERK1/2 was reduced and p-p38 was increased. Proliferation of porcine skeletal muscle cells was induced after GPX2 overexpression, was accompanied by activation in JNK, ERK1/2, and p-p38. Overexpression methods confirmed that GPX2 has a promoting function in myoblastic differentiation. ERK1/2 pathway was activated and p38 was suppressed during the process. This study lays a foundation for the functional study of GPX2 and provides theoretical support for promoting subcutaneous fat reduction and muscle growth.


Assuntos
Adipócitos , Glutationa Peroxidase , Sistema de Sinalização das MAP Quinases , Animais , Glutationa Peroxidase/metabolismo , Glutationa Peroxidase/genética , Adipócitos/metabolismo , Adipócitos/citologia , Suínos , Diferenciação Celular/genética , Proliferação de Células , Adipogenia/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/citologia , Músculo Esquelético/metabolismo , Músculo Esquelético/citologia
11.
Cells ; 13(9)2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38727299

RESUMO

The adipose organ adapts and responds to internal and environmental stimuli by remodeling both its cellular and extracellular components. Under conditions of energy surplus, the subcutaneous white adipose tissue (WAT) is capable of expanding through the enlargement of existing adipocytes (hypertrophy), followed by de novo adipogenesis (hyperplasia), which is impaired in hypertrophic obesity. However, an impaired hyperplastic response may result from various defects in adipogenesis, leading to different WAT features and metabolic consequences, as discussed here by reviewing the results of the studies in animal models with either overexpression or knockdown of the main molecular regulators of the two steps of the adipogenesis process. Moreover, impaired WAT remodeling with aging has been associated with various age-related conditions and reduced lifespan expectancy. Here, we delve into the latest advancements in comprehending the molecular and cellular processes underlying age-related changes in WAT function, their involvement in common aging pathologies, and their potential as therapeutic targets to influence both the health of elderly people and longevity. Overall, this review aims to encourage research on the mechanisms of WAT maladaptation common to conditions of both excessive and insufficient fat tissue. The goal is to devise adipocyte-targeted therapies that are effective against both obesity- and age-related disorders.


Assuntos
Adipogenia , Tecido Adiposo Branco , Envelhecimento , Obesidade , Humanos , Envelhecimento/patologia , Obesidade/patologia , Obesidade/metabolismo , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/patologia , Animais , Adipócitos/metabolismo , Adipócitos/patologia
12.
Dev Cell ; 59(10): 1231-1232, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38772342

RESUMO

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.


Assuntos
Adipogenia , Receptores Notch , Células-Tronco , Adipogenia/fisiologia , Animais , Receptores Notch/metabolismo , Células-Tronco/metabolismo , Células-Tronco/citologia , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Marrom/citologia , Humanos , Adipócitos Marrons/metabolismo , Adipócitos Marrons/citologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Diferenciação Celular , Linhagem da Célula , Camundongos , Transdução de Sinais
13.
Front Endocrinol (Lausanne) ; 15: 1385811, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38765953

RESUMO

Background: Thermogenic beige adipocytes, which dissipate energy as heat, are found in neonates and adults. Recent studies show that neonatal beige adipocytes are highly plastic and contribute to >50% of beige adipocytes in adults. Neonatal beige adipocytes are distinct from recruited beige adipocytes in that they develop independently of temperature and sympathetic innervation through poorly defined mechanisms. Methods: We characterized the neonatal beige adipocytes in the inguinal white adipose tissue (iWAT) of C57BL6 postnatal day 3 and 20 mice (P3 and P20) by imaging, genome-wide RNA-seq analysis, ChIP-seq analysis, qRT-PCR validation, and biochemical assays. Results: We found an increase in acetylated histone 3 lysine 27 (H3K27ac) on the promoter and enhancer regions of beige-specific gene UCP1 in iWAT of P20 mice. Furthermore, H3K27ac ChIP-seq analysis in the iWAT of P3 and P20 mice revealed strong H3K27ac signals at beige adipocyte-associated genes in the iWAT of P20 mice. The integration of H3K27ac ChIP-seq and RNA-seq analysis in the iWAT of P20 mice reveal epigenetically active signatures of beige adipocytes, including oxidative phosphorylation and mitochondrial metabolism. We identify the enrichment of GA-binding protein alpha (GABPα) binding regions in the epigenetically active chromatin regions of the P20 iWAT, particularly on beige genes, and demonstrate that GABPα is required for beige adipocyte differentiation. Moreover, transcriptomic analysis and glucose oxidation assays revealed increased glycolytic activity in the neonatal iWAT from P20. Conclusions: Our findings demonstrate that epigenetic mechanisms regulate the development of peri-weaning beige adipocytes via GABPα. Further studies to better understand the upstream mechanisms that regulate epigenetic activation of GABPα and characterization of the metabolic identity of neonatal beige adipocytes will help us harness their therapeutic potential in metabolic diseases.


Assuntos
Adipócitos Bege , Adipogenia , Tecido Adiposo Branco , Animais Recém-Nascidos , Cromatina , Epigênese Genética , Fator de Transcrição de Proteínas de Ligação GA , Camundongos Endogâmicos C57BL , Animais , Camundongos , Adipócitos Bege/metabolismo , Cromatina/metabolismo , Cromatina/genética , Adipogenia/genética , Tecido Adiposo Branco/metabolismo , Fator de Transcrição de Proteínas de Ligação GA/metabolismo , Fator de Transcrição de Proteínas de Ligação GA/genética , Masculino , Termogênese/genética , Histonas/metabolismo , Histonas/genética
14.
J Endocrinol ; 262(1)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38692289

RESUMO

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.


Assuntos
Células 3T3-L1 , Adipócitos , Adipogenia , Ciclo Celular , Receptores de Hialuronatos , PPAR gama , Adipogenia/genética , Adipogenia/fisiologia , Receptores de Hialuronatos/metabolismo , Receptores de Hialuronatos/genética , Animais , PPAR gama/metabolismo , PPAR gama/genética , Camundongos , Ciclo Celular/genética , Ciclo Celular/fisiologia , Humanos , Adipócitos/metabolismo , Deleção de Genes , Diferenciação Celular/genética , Masculino , Tecido Adiposo/metabolismo , Tecido Adiposo/citologia , Transdução de Sinais/fisiologia
15.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 38(5): 626-632, 2024 May 15.
Artigo em Chinês | MEDLINE | ID: mdl-38752252

RESUMO

Objective: To summarize the dynamic and synchronized changes between the hair cycle and dermal adipose tissue as well as the impact of dermal adipose tissue on hair growth, and to provide a new research idea for the clinical treatment of hair loss. Methods: An extensive review of relevant literature both domestic and international was conducted, analyzing and summarizing the impact of dermal adipose precursor cells, mature dermal adipocytes, and the processes of adipogenesis in dermal adipose tissue on the transition of hair cycle phases. Results: Dermal adipose tissue is anatomically adjacent to hair follicles and closely related to the changes in the hair cycle. The proliferation and differentiation of dermal adipose precursor cells promote the transition of hair cycle from telogen to anagen, while mature adipocytes can accelerate the transition from anagen to catagen of the hair cycle by expressing signaling molecules, with adipogenesis in dermal adipose tissue and hair cycle transition signaling coexistence. Conclusion: Dermal adipose tissue affects the transition of the hair cycle and regulates hair growth by secreting various signaling molecules. However, the quantity and depth of existing literature are far from sufficient to fully elucidate its prominent role in regulating the hair cycle, and the specific regulatory mechanisms needs to be further studied.


Assuntos
Adipócitos , Adipogenia , Tecido Adiposo , Diferenciação Celular , Folículo Piloso , Cabelo , Humanos , Tecido Adiposo/metabolismo , Tecido Adiposo/citologia , Folículo Piloso/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Cabelo/crescimento & desenvolvimento , Cabelo/metabolismo , Transdução de Sinais , Derme/metabolismo , Derme/citologia , Animais , Proliferação de Células , Alopecia/metabolismo
16.
FASEB J ; 38(9): e23657, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38713087

RESUMO

The pathogenesis of osteoporosis (OP) is closely associated with the disrupted balance between osteogenesis and adipogenesis in bone marrow-derived mesenchymal stem cells (BMSCs). We analyzed published single-cell RNA sequencing (scRNA-seq) data to dissect the transcriptomic profiles of bone marrow-derived cells in OP, reviewing 56 377 cells across eight scRNA-seq datasets from femoral heads (osteoporosis or osteopenia n = 5, osteoarthritis n = 3). Seventeen genes, including carboxypeptidase M (CPM), were identified as key osteogenesis-adipogenesis regulators through comprehensive gene set enrichment, differential expression, regulon activity, and pseudotime analyses. In vitro, CPM knockdown reduced osteogenesis and promoted adipogenesis in BMSCs, while adenovirus-mediated CPM overexpression had the reverse effects. In vivo, intraosseous injection of CPM-overexpressing BMSCs mitigated bone loss in ovariectomized mice. Integrated scRNA-seq and bulk RNA sequencing analyses provided insight into the MAPK/ERK pathway's role in the CPM-mediated regulation of BMSC osteogenesis and adipogenesis; specifically, CPM overexpression enhanced MAPK/ERK signaling and osteogenesis. In contrast, the ERK1/2 inhibitor binimetinib negated the effects of CPM overexpression. Overall, our findings identify CPM as a pivotal regulator of BMSC differentiation, which provides new clues for the mechanistic study of OP.


Assuntos
Adipogenia , Sistema de Sinalização das MAP Quinases , Células-Tronco Mesenquimais , Metaloendopeptidases , Osteogênese , Análise de Célula Única , Animais , Osteogênese/fisiologia , Osteogênese/genética , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Feminino , Transcriptoma , Carboxipeptidases/metabolismo , Carboxipeptidases/genética , Humanos , Diferenciação Celular , Osteoporose/genética , Osteoporose/metabolismo , Osteoporose/patologia , Camundongos Endogâmicos C57BL , Proteínas Ligadas por GPI
17.
Mol Genet Genomics ; 299(1): 48, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38700639

RESUMO

Intramuscular fat (IMF) is a critical factor in beef quality. IMF is mainly distributed between muscle fibres and its accumulation can affect the marbling and meat quality of beef. IMF formation and deposition is a complex process and in recent years a group of non-coding RNAs (ncRNAs), known as circRNAs, have been discovered to play an important role in regulating intramuscular fat deposition. CircRNAs form a covalent loop structure after reverse splicing of precursor mRNAs. They can act by adsorbing miRNAs, thereby reducing their repressive effects on downstream target genes. Based on high-throughput sequencing of circRNAs in intramuscular fat of Qinchuan and Japanese black cattle, we identified a novel circSSBP2 that is differentially expressed between the two species and associated with adipogenesis. We show that circSSBP2 knockdown promotes bovine intramuscular preadipocyte proliferation, whereas overexpression inhibits bovine intramuscular preadipocyte proliferation. We also show that circSSBP2 can act as a molecular sponge for miR-2400 and that miR-2400 overexpression promotes bovine intramuscular preadipocyte proliferation. Furthermore, N-myc downstream-regulated gene 1 (NDRG1) was identified as a direct target gene of miR-2400, and NDRG1 interference promoted the proliferation of bovine intramuscular preadipocytes. In conclusion, our results suggest that circSSBP2 inhibits the proliferation of bovine intramuscular preadipocytes by regulating the miR-2400/NDRG1 axis.


Assuntos
Adipócitos , Adipogenia , Proteínas de Ciclo Celular , Proliferação de Células , Peptídeos e Proteínas de Sinalização Intracelular , MicroRNAs , Animais , MicroRNAs/genética , MicroRNAs/metabolismo , Proliferação de Células/genética , Bovinos , Adipócitos/metabolismo , Adipócitos/citologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Adipogenia/genética , RNA Circular/genética , Regulação da Expressão Gênica
18.
Int J Mol Sci ; 25(9)2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38732125

RESUMO

"Ganghwal" is a widely used herbal medicine in Republic of Korea, but it has not been reported as a treatment strategy for obesity and diabetes within adipocytes. In this study, we determined that Ostericum koreanum extract (OKE) exerts an anti-obesity effect by inhibiting adipogenesis and an anti-diabetic effect by increasing the expression of genes related to glucose uptake in adipocytes and inhibiting α-glucosidase activity. 3T3-L1 preadipocytes were differentiated for 8 days in methylisobutylxanthine, dexamethasone, and insulin medium, and the effect of OKE was confirmed by the addition of 50 and 100 µg/mL of OKE during the differentiation process. This resulted in a reduction in lipid accumulation and the expression of PPARγ (Peroxisome proliferator-activated receptor γ) and C/EBPα (CCAAT enhancer binding protein α). Significant activation of AMPK (AMP-activated protein kinase), increased expression of GLUT4 (Glucose Transporter Type 4), and inhibition of α-glucosidase activity were also observed. These findings provide the basis for the anti-obesity and anti-diabetic effects of OKE. In addition, OKE has a significant antioxidant effect. This study presents OKE as a potential natural product-derived material for the treatment of patients with metabolic diseases such as obesity- and obesity-induced diabetes.


Assuntos
Células 3T3-L1 , Adipócitos , Adipogenia , Fármacos Antiobesidade , Hipoglicemiantes , PPAR gama , Extratos Vegetais , Camundongos , Extratos Vegetais/farmacologia , Extratos Vegetais/química , Animais , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Adipogenia/efeitos dos fármacos , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , PPAR gama/metabolismo , PPAR gama/genética , Fármacos Antiobesidade/farmacologia , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Transportador de Glucose Tipo 4/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Proteína alfa Estimuladora de Ligação a CCAAT/genética , alfa-Glucosidases/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Antioxidantes/farmacologia , Inibidores de Glicosídeo Hidrolases/farmacologia , Crassulaceae/química , Metabolismo dos Lipídeos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos
19.
Sci Rep ; 14(1): 10924, 2024 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-38740866

RESUMO

Bovine intramuscular fat (IMF), commonly referred to as marbling, is regulated by lipid metabolism, which includes adipogenesis, lipogenesis, glycerolipid synthesis, and lipolysis. In recent years, breeding researchers have identified single nucleotide polymorphisms (SNPs) as useful marker-assisted selection tools for improving marbling scores in national breeding programs. These included causal SNPs that induce phenotypic variation. MicroRNAs (miRNAs) are small highly conserved non-coding RNA molecules that bind to multiple non-coding regions. They are involved in post-transcriptional regulation. Multiple miRNAs may regulate a given target. Previously, three SNPs in the GPAM 3' UTR and four miRNAs were identified through in silico assays. The aim of this study is to verify the binding ability of the four miRNAs to the SNPs within the 3'UTR of GPAM, and to identify the regulatory function of miR-375 in the expression of genes related to lipid metabolism in mammalian adipocytes. It was verified that the four miRNAs bind to the GPAM 3'UTR, and identified that the miR-375 sequence is highly conserved. Furthermore, it was founded that miR-375 upregulated the GPAM gene, C/EBPα, PPARγ and lipid metabolism-related genes and promoted lipid droplet accumulation in 3T3-L1 cells. In conclusion, these results suggest that miR-375 is a multifunctional regulator of multiple lipid metabolism-related genes and may aid in obesity research as a biomarker.


Assuntos
Regiões 3' não Traduzidas , Células 3T3-L1 , Metabolismo dos Lipídeos , MicroRNAs , Polimorfismo de Nucleotídeo Único , MicroRNAs/genética , MicroRNAs/metabolismo , Animais , Camundongos , Metabolismo dos Lipídeos/genética , Bovinos , Regulação da Expressão Gênica , Adipócitos/metabolismo , Adipogenia/genética
20.
Curr Protoc ; 4(4): e1032, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38606955

RESUMO

The therapeutic potential of mesenchymal stromal cells (MSCs) has been extensively investigated in both preclinical and clinical settings. Recent years have witnessed the emergence of numerous isolation protocols and culture techniques, ranging from the selection of subpopulations to preserve stemness to preconditioning strategies aimed at enhancing therapeutic efficacy, tailored to the specific tissue source. In this protocol, we present a straightforward and cost-effective method for isolating human MSCs (hMSCs) from discarded bone marrow collection kits (comprising bag and filter systems) originally intended for removing impurities and unwanted cellular debris from the collected bone marrow aspirate, ensuring the purity of the stem cell population during stem cell transplantation. Utilizing basic laboratory equipment, we demonstrate the isolation of hMSCs, highlighting the expression of specific surface antigens, and multilineage differentiation into adipogenic, osteogenic, and chondrogenic lineages in vitro. This sustainable and resource-efficient approach not only contributes to reducing medical waste but also holds promise for advancing regenerative medicine applications. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Isolation of human mesenchymal stromal cells from bone marrow collection kits Basic Protocol 2: Culture of human mesenchymal stromal cells Basic Protocol 3: Characterization of human mesenchymal stromal cells with flow cytometry analysis Basic Protocol 4: Characterization of human mesenchymal stromal cells with multilineage differentiation under in vitro conditions.


Assuntos
Medula Óssea , Células-Tronco Mesenquimais , Humanos , Células-Tronco Mesenquimais/metabolismo , Diferenciação Celular , Adipogenia , Citometria de Fluxo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA