Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 15.442
Filtrar
1.
Adipocyte ; 13(1): 2421750, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39484712

RESUMO

Reciprocal communication between adipocytes and immune cells is essential to maintain optimal adipose tissue (AT) functionality. Amongst others, adipocytes directly interact with invariant NKT cells (iNKT cells), which in turn secrete various cytokines. A lipid-rich microenvironment, as observed in obesity, skews this adipocyte-driven cytokine output towards a more inflammatory output. Whether a lipid-rich microenvironment also affects iNKT cells directly, however, is unknown. Here, we show that primary mouse iNKT cells isolated from AT can accumulate lipids in lipid droplets (LDs), more so than liver- and spleen-resident iNKT cells. Furthermore, a lipid-rich microenvironment increased the production of the proinflammatory cytokine IFNγ. Next, to an indirect, adipocyte-mediated cue, iNKT cells can directly respond to environmental lipid changes, supporting a potential role as nutrient sensors.


Assuntos
Tecido Adiposo , Inflamação , Metabolismo dos Lipídeos , Células T Matadoras Naturais , Animais , Células T Matadoras Naturais/metabolismo , Células T Matadoras Naturais/imunologia , Camundongos , Tecido Adiposo/metabolismo , Inflamação/metabolismo , Fenótipo , Camundongos Endogâmicos C57BL , Adipócitos/metabolismo , Gotículas Lipídicas/metabolismo , Masculino , Citocinas/metabolismo , Interferon gama/metabolismo
2.
Cancer Med ; 13(20): e70265, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39431622

RESUMO

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer. Recent studies indicated that cancer-associated adipocytes (CAAs) play crucial roles in tumor progression; however, the precise mechanism remains unknown. Here, we analyzed specific exosomal microRNAs (miRNA) signatures derived from pancreatic CAAs to investigate their role in cancer progression. METHODS: CAAs were generated by co-culturing human adipocytes with human pancreatic cancer cells, and exosomes were isolated from the CAA-conditioned medium (CAA-CM). Small RNA-seq analysis was used to identify differentially expressed miRNAs in these exosomes. The effects of miRNAs on cell proliferation, migration/invasion, and drug sensitivity were examined. Luciferase reporter assays, real-time polymerase chain reaction, and western blotting were performed to investigate the molecular mechanisms of the miRNAs. The clinical relevance of the miRNAs was investigated using publicly available data and our cohort of patients with PDAC. RESULTS: miR-199a-3p expression was significantly increased in CAA-CM-derived exosomes. CAA-derived exosomes transferred miR-199a-3p to pancreatic cancer cells. Transfection with miR-199a-3p increased the proliferation, invasion, migration, and drug resistance of pancreatic cancer cells by downregulating SOCS7, increasing STAT3 phosphorylation, and upregulating SAA1 expression. High tissue miR-199a-3p expression is correlated with poor prognosis in patients with PDAC. Liquid biopsies revealed that exosomal miR-199a-3p could accurately differentiate patients with PDAC from healthy controls. Multivariate survival analysis indicated that miR-199a is an independent prognostic factor for PDAC. CONCLUSION: miR-199a-3p in CAA-derived exosomes contributes to the malignant transformation of pancreatic cancer via the SOCS7/STAT3/SAA1 pathway, suggesting its potential as a biomarker and therapeutic target for PDAC.


Assuntos
Adipócitos , Carcinoma Ductal Pancreático , Proliferação de Células , Progressão da Doença , Exossomos , Regulação Neoplásica da Expressão Gênica , MicroRNAs , Neoplasias Pancreáticas , Humanos , MicroRNAs/genética , Exossomos/metabolismo , Exossomos/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/metabolismo , Adipócitos/metabolismo , Adipócitos/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/genética , Prognóstico , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Feminino , Masculino , Resistencia a Medicamentos Antineoplásicos/genética
3.
FASEB J ; 38(19): e70093, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39373976

RESUMO

The risk of developing type 2 diabetes (T2D) is heterogeneous among individuals with obesity. Functional decline of adipocyte precursor cells (APCs) and accumulation of senescent cells in the subcutaneous adipose tissue contributes to the progression toward T2D. LncRNAs regulate cell senescence and may be implicated in determining this abnormality in APCs. Here, we report that APCs from individuals with obesity show a gradual increase in multiple senescence markers, which worsens in parallel with the progression from normal glucose tolerance (NGT) to impaired glucose tolerance (IGT) or T2D. Transcriptomic analysis identified PANDAR as the top-ranked lncRNA differentially expressed in APCs from individuals with obesity and T2D and non-obese subjects. Q-PCR confirmed PANDAR up-regulation in APCs from individuals with obesity, at progressively increased levels in those who developed, respectively, IGT and T2D. Bisulfite sequencing and luciferase assays revealed that, in parallel with glucose tolerance deterioration, the -1317 CpG at the PANDAR promoter became hypo-methylated in obesity, resulting in enhanced PANDAR induction by p53. PANDAR silencing in senescent APCs from individuals with obesity and T2D caused repression of senescence programs and cell cycle re-entry. PANDAR transcription in white blood cells (WBCs) mirrored that in APCs. Also, individuals with obesity exhibited rescue of PANDAR transcription in WBCs following bariatric surgery, accompanied by enhanced methylation at the regulatory PANDAR -1317 CpG. In conclusion, PANDAR dysregulation is a newly identified mechanism determining the early senescence of APCs from individuals with obesity, which worsens along the progression toward T2D. In the future, PANDAR targeting may represent a valuable strategy to delay this progression.


Assuntos
Adipócitos , Senescência Celular , Metilação de DNA , Diabetes Mellitus Tipo 2 , Obesidade , Regiões Promotoras Genéticas , RNA Longo não Codificante , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adipócitos/metabolismo , Senescência Celular/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Obesidade/genética , Obesidade/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
4.
J Biosci ; 492024.
Artigo em Inglês | MEDLINE | ID: mdl-39377444

RESUMO

The regulation of adipose tissue metabolism by irisin involves modulating gene expressions related to energy metabolism and insulin sensitivity via miRNA-mediated signaling pathways within adipose tissue. Understanding the molecular mechanisms behind the role of irisin is vital for addressing obesity and related metabolic complications. In this study, we undertook an extensive miRNA transcriptomic approach to identify differentially expressed miRNAs following irisin exposure in adipocytes and murine white adipose tissue. Our findings spotlighted two miRNAs, miRNA-758 and miRNA-668, as being influenced by irisin. To understand the impact of the modulations of these miRNAs by irisin, we performed a signaling pathway and network analysis. After irisin exposure, both, miRNA-758 and miRNA-668, emerged as key regulators in leptin and CDK5 signaling pathways. Leptin, a hormone originating from adipose tissue, is primarily produced by adipocytes, and its effects are known to be mediated by CDK5. In essence, this study identifies pivotal genes and miRNAs in irisin-driven mechanisms in adipose tissue, offering valuable insights for crafting novel therapeutic strategies for metabolic and associated disorders.


Assuntos
Adipócitos , Quinase 5 Dependente de Ciclina , Fibronectinas , MicroRNAs , Transdução de Sinais , MicroRNAs/genética , MicroRNAs/metabolismo , Fibronectinas/metabolismo , Fibronectinas/genética , Animais , Camundongos , Adipócitos/metabolismo , Quinase 5 Dependente de Ciclina/metabolismo , Quinase 5 Dependente de Ciclina/genética , Leptina/metabolismo , Leptina/genética , Regulação da Expressão Gênica , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/metabolismo , Células 3T3-L1 , Humanos , Perfilação da Expressão Gênica , Metabolismo Energético/genética , Obesidade/metabolismo , Obesidade/genética
5.
Adipocyte ; 13(1): 2411453, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39385585

RESUMO

Obesity is a globally prevalent metabolic disorder characterized by an increased number of adipose cells and excessive fat in adipocytes. Herbal medicines, such as ginger, have shown potential in treating obesity by inhibiting adipogenesis and reducing adipocyte hypertrophy. Ginger contains bioactive compounds, particularly gingerols, which have demonstrated anti-adipogenic and/or lipolytic effects. However, research on the effects of 10-gingerol on adipose tissue remains limited. This study aimed to evaluate the effect of 10-gingerol on lipid content, lipolysis markers, and the expression of genes related to lipid metabolism in 3T3-L1 adipocytes. Three groups were analyzed: a negative control (preadipocytes), a positive control (mature adipocytes), and a group treated with 10-gingerol (10-G). Results showed that 10-G reduced lipid accumulation by 42.16% in mature adipocytes compared to the control, without affecting cell viability. Additionally, 10-G increased glycerol release and downregulated lipogenic genes such as Pparγ, Acaca, Fabp4, and Mtor, while upregulating genes related to fatty acid oxidation, including Cebpα, Cpt1a, Lipe, and Prkaa1. In conclusion, 10-gingerol reduces lipid content in mature adipocytes by downregulating lipogenesis, increasing lipolysis, and enhancing fatty acid oxidation.


Assuntos
Células 3T3-L1 , Adipócitos , Catecóis , Álcoois Graxos , Gotículas Lipídicas , Metabolismo dos Lipídeos , Lipólise , Animais , Álcoois Graxos/farmacologia , Lipólise/efeitos dos fármacos , Catecóis/farmacologia , Camundongos , Adipócitos/metabolismo , Adipócitos/efeitos dos fármacos , Gotículas Lipídicas/metabolismo , Gotículas Lipídicas/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Lipogênese/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos
6.
Results Probl Cell Differ ; 74: 159-174, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39406905

RESUMO

In obesity, adipose tissue macrophages (ATMs) are abundant immune cells in the adipose tissue and are known as inducers of metabolic inflammation that may lead to insulin resistance and immune disorders associated with obesity. However, much less is known about the ontogeny and physiological functions of ATMs in lean adipose tissue. ATMs are present at birth and actively participate in the synthesis of mediators that induce lipolysis, mitobiogenesis, and thermogenesis in adipocytes. Later in life ATMs limit the thermogenic competence of the adipocytes and favor lipid storage. ATMs respond to lipid overload of adipocytes in obesity with a sequence of pro-inflammatory events, including inflammasome activation and pyroptosis, as well as stimulation of nuclear factor kappa B and interferon regulatory factors that evoke an uncontrolled inflammation. ATMs are life-long constituents of the adipose tissue and hence signals that control ATM development and ATM-adipocyte interactions determine adipose tissue health.


Assuntos
Tecido Adiposo , Macrófagos , Humanos , Tecido Adiposo/metabolismo , Macrófagos/metabolismo , Animais , Obesidade/metabolismo , Adipócitos/metabolismo , Inflamação
7.
Adipocyte ; 13(1): 2414919, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39415617

RESUMO

Tumor necrosis factor alpha (TNF-α)/hypoxia-treated 3T3-L1 adipocytes have been used to model inflamed and insulin-resistant adipose tissue: this study examines gaps in the model. We tested whether modulating TNF-α/hypoxia treatment time could reduce cell death while still inducing inflammation and insulin resistance. Adipocytes were treated with TNF-α (12 h or 24 h) and incubated in a hypoxic chamber for 24 h. To examine maintenance of the phenotype over time, glucose and FBS were added at 24 h post initiation of treatment, and the cells were maintained for an additional 48 h. Untreated adipocytes were used as a control. Viability, insulin resistance, and inflammation were assessed using Live/Dead staining, RT-qPCR, ELISA, and glucose uptake assays. Treatment for 12 h with TNF-α in the presence of hypoxia resulted in an increase in the percentage of live cells compared to 24 h treated cells. Importantly, insulin resistance and inflammation were still induced in the 12 h treated adipocytes: the expression of the insulin sensitive and inflammatory genes was decreased and increased, respectively. In 72 h treated adipocytes, no significant differences were found in the viability, glucose uptake or insulin-sensitive and inflammatory gene expression. This study provides a modified approach to in vitro odeling adipocyte inflammation and insulin resistance. .


Assuntos
Células 3T3-L1 , Adipócitos , Inflamação , Resistência à Insulina , Fator de Necrose Tumoral alfa , Animais , Camundongos , Adipócitos/metabolismo , Inflamação/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Glucose/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Hipóxia Celular
8.
Sci Rep ; 14(1): 23920, 2024 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-39397046

RESUMO

Our research explores the detrimental effects of microplastic (MP) exposure on adipose tissue aging and function, emphasizing the potential health risks associated with environmental pollutants. Utilizing both in vivo and in vitro models, we discovered that MPs accumulate in adipose tissues, leading to cellular senescence, inflammation, and hindered adipogenic differentiation. Notably, our findings demonstrate that MPs prompt an aging response in both epididymal and inguinal white adipose tissue, increase senescence-associated ß-galactosidase activity, and upregulate key senescence and inflammatory markers. Furthermore, we show that MPs disrupt normal adipogenic differentiation by reducing lipid droplet formation and downregulating critical adipogenic markers. These insights highlight the urgent need for further investigation into the long-term consequences of MP pollution on biological aging and underscore the importance of developing public health strategies to mitigate these effects.


Assuntos
Adipócitos , Adipogenia , Senescência Celular , Microplásticos , Adipogenia/efeitos dos fármacos , Animais , Microplásticos/toxicidade , Senescência Celular/efeitos dos fármacos , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Camundongos , Masculino , Diferenciação Celular/efeitos dos fármacos , Envelhecimento/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/efeitos dos fármacos , Camundongos Endogâmicos C57BL
9.
Int J Mol Sci ; 25(19)2024 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-39409054

RESUMO

Inflammatory Bowel Diseases (IBDs) are associated with aberrant immune function, widespread inflammation, and altered intestinal blood flow. Perivascular adipose tissue (PVAT) surrounding the mesenteric vasculature can modulate vascular function and control the local immune cell population, but its structure and function have never been investigated in IBD. We used an IL10-/- mouse model of colitis that shares features with human IBD to test the hypothesis that IBD is associated with (1) impaired ability of PVAT to dilate mesenteric arteries and (2) changes in PVAT resident adipocyte and immune cell populations. Pressure myography and electrical field stimulation of isolated mesenteric arteries show that PVAT not only loses its anti-contractile effect but becomes pro-contractile in IBD. Quantitative immunohistochemistry and confocal imaging studies found significant adipocyte hyperplasia and increased PVAT leukocytes, particularly macrophages, in IBD. PCR arrays suggest that these changes occur alongside the altered cytokine and chemokine gene expression associated with altered NF-κB signaling. Collectively, these results show that the accumulation of macrophages in PVAT during IBD pathogenesis may lead to local inflammation, which ultimately contributes to increased arterial constriction and decreased intestinal blood flow with IBD.


Assuntos
Tecido Adiposo , Colite , Modelos Animais de Doenças , Doenças Inflamatórias Intestinais , Interleucina-10 , Animais , Camundongos , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Doenças Inflamatórias Intestinais/genética , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Interleucina-10/metabolismo , Interleucina-10/genética , Colite/metabolismo , Colite/patologia , Colite/genética , Macrófagos/metabolismo , Artérias Mesentéricas/metabolismo , Artérias Mesentéricas/patologia , Camundongos Knockout , Camundongos Endogâmicos C57BL , Masculino , NF-kappa B/metabolismo , Adipócitos/metabolismo , Adipócitos/patologia , Citocinas/metabolismo
10.
Nutrients ; 16(19)2024 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-39408236

RESUMO

BACKGROUND: Rosa canina L. (rosehip) is used worldwide in traditional medicine as a plant with medicinal properties. However, its anti-obesity effects are not fully explained on a transcriptional level. METHODS: In the present work, the 3T3-L preadipocytes were utilized to explore the impact of R. canina fruit extract (RCE) on the cellular and molecular pathways involved in adipocyte hypertrophy. RESULTS: Obtained results showed the ability of RCE to reduce lipid overloads in hypertrophic adipocytes associated with the down-regulation of mRNA expressions of adipogenic transcription factors such as PPARγ, C/EBPα, and SREBP-1c as well as genes involved in lipid biosyntheses such as FAS, LPL, and aP2. Moreover, obesity-associated oxidative stress (antioxidant enzyme activities and ROS generation) and inflammation were ameliorated in RCE-treated hypertrophic adipocytes. The mRNA and protein levels of adipokines such as leptin, resistin, and adiponectin were restored to more favorable levels. CONCLUSIONS: Rosa canina fruit might be a valuable source of phytochemicals in preventing obesity and obesity-related metabolic complications.


Assuntos
Células 3T3-L1 , Adipócitos , Metabolismo dos Lipídeos , Extratos Vegetais , Espécies Reativas de Oxigênio , Rosa , Animais , Rosa/química , Camundongos , Extratos Vegetais/farmacologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Inflamação/metabolismo , Inflamação/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Frutas/química , PPAR gama/metabolismo , Obesidade/metabolismo , Hipertrofia , Fármacos Antiobesidade/farmacologia , Adipocinas/metabolismo
11.
Int J Med Sci ; 21(13): 2480-2493, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39439458

RESUMO

Background: Adipocytes play a crucial role in tissue regeneration, contributing to the restoration of damaged areas and modulating the inflammatory milieu. The modulation of gene expression through chemically modified PPARγ mRNA (PPARγ-modRNA) introduces a sophisticated approach to precisely control adipogenic processes. This study aims to explore the adipogenic potential of the PPARγ-modRNA in 3T3-L1 preadipocytes and its role in wound healing. Materials and Methods: We transfected 3T3-L1 preadipocytes with PPARγ-modRNA to investigate adipogenic differentiation and cellular proliferation in vitro. In vivo, we employed a murine full-thickness skin defect model and compared the effects of modRNA-mediated PPARγ overexpression with control groups. Additionally, we conducted RNA sequencing on luciferase-modified mRNA (LUC) and PPARγ-modRNA-transfected cells (PPAR) for a comprehensive understanding of molecular mechanisms. Results: PPARγ-modRNA significantly enhanced adipogenesis and proliferation in 3T3-L1 preadipocytes in vitro. The injection of PPARγ-modified mRNA led to accelerated wound healing compared to the control groups in vivo. RNA sequencing revealed upregulation of adipogenesis-related genes in the PPAR group, notably associated with the TNF signaling pathway. Subsequently, the KEGG analysis indicated that modRNA-mediated PPARγ overexpression effectively promoted adipogenesis while inhibiting TNF-α-mediated inflammation and cellular apoptosis. Conclusions: This study demonstrates the innovative use of PPARγ-modRNA to induce adipogenesis and expedite wound healing. The nuclear expression of PPARγ through modRNA technology signifies a notable advancement, with implications for future therapeutic strategies targeting adipogenic processes and the inhibition of inflammation in the context of wound healing.


Assuntos
Células 3T3-L1 , Adipócitos , Adipogenia , Proliferação de Células , PPAR gama , RNA Mensageiro , Cicatrização , Animais , PPAR gama/metabolismo , PPAR gama/genética , Camundongos , Adipogenia/genética , Cicatrização/genética , Adipócitos/metabolismo , Adipócitos/citologia , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Proliferação de Células/genética , Diferenciação Celular/genética , Humanos , Modelos Animais de Doenças , Pele/patologia , Pele/metabolismo , Masculino
12.
Nat Commun ; 15(1): 9107, 2024 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-39438462

RESUMO

Cultured or cultivated meat, animal muscle, and fat tissue grown in vitro, could transform the global meat market, reducing animal suffering while using fewer resources than traditional meat production and no antimicrobials at all. To ensure the appeal of cultured meat to future customers, cultured fat is essential for achieving desired mouthfeel, taste, and texture, especially in beef. In this work we show the establishment of primary bovine adipose-derived stem cell spheroids in static and dynamic suspension culture. Spheroids are successfully differentiated using a single-step protocol. Differentiated spheroids from dynamic cultures maintain stability and viability during 3D bioprinting in edible gellan gum. Also, the fatty acid composition of differentiated spheroids is significantly different from control spheroids. The cells are cultured antibiotic-free to minimize the use of harmful substances. This work presents a stable and bioprintable building block for cultured fat with a high cell density in a 3D dynamic cell culture system.


Assuntos
Tecido Adiposo , Esferoides Celulares , Células-Tronco , Animais , Bovinos , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Diferenciação Celular , Engenharia Tecidual/métodos , Células Cultivadas , Técnicas de Cultura de Células/métodos , Bioimpressão/métodos , Adipócitos/citologia , Adipócitos/metabolismo , Ácidos Graxos/metabolismo
13.
Yi Chuan ; 46(10): 860-870, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39443314

RESUMO

GULP1 is an engulfment adaptor protein containing a phosphotyrosine-binding (PTB) domain, and existing studies have shown that it can promote glucose uptake in 3T3-L1 adipocytes. To further explore key metabolically related differential genes downstream of GULP1, this study conducted transcriptome analysis on adipocytes and skeletal muscle cells overexpressing GULP1. Subsequently, abnormally expressed genes were subjected to bioinformatic analysis, and real-time fluorescent quantitative PCR (qRT-PCR) was used for mutual validation with transcriptome sequencing. The results indicated that, with a threshold of P < 0.05 and |Log2FoldChange| ≥ 1 for screening differentially expressed genes, compared with control cells, there were 278 upregulated and 263 downregulated genes in adipocytes overexpressing GULP1. Metabolism-related GO (Gene Ontology) terms included cholesterol biosynthetic process, cholesterol metabolic process, response to lipopolysaccharide, lipid metabolic process, etc. A total of 52 metabolically related differentially expressed genes were enriched in 10 KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways, with lipid metabolism being highly enriched. In skeletal muscle cells overexpressing GULP1, there were 280 upregulated and 302 downregulated genes, with metabolism-related GO terms including hormone metabolic process, response to lipopolysaccharide, one-carbon metabolic process, etc. A total of 86 metabolically related differentially expressed genes were enriched in 10 KEGG pathways, with amino acid metabolism, lipid metabolism, and carbohydrate metabolism being highly enriched. GULP1's biological functions are extensive, including lipid metabolism and oncology. This study, through transcriptomics and bioinformatic analysis, identified key metabolically related differential genes downstream of GULP1, obtained metabolically related differential genes and signaling pathways after GULP1 overexpression, providing important theoretical basis for future research on GULP1 downstream target genes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Adipócitos , Animais , Camundongos , Células 3T3-L1 , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adipócitos/metabolismo , Perfilação da Expressão Gênica/métodos , Metabolismo dos Lipídeos/genética , Transcriptoma
14.
Front Endocrinol (Lausanne) ; 15: 1468824, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39444451

RESUMO

Background: Studying the molecular mechanisms of lipodystrophy can provide valuable insights into the pathophysiology of insulin resistance (IR), type 2 diabetes (T2D), and other clustering diseases [metabolic syndrome (MetS)] and its underlying adipocentric disease (MetS disease). Methods: A high-confidence lipodystrophy gene panel comprising 50 genes was created, and their expressions were measured in the visceral and subcutaneous (both peripheral and abdominal) adipose depots of MetS and non-MetS individuals at a tertiary care medical facility. Results: Most lipodystrophy genes showed significant downregulation in MetS individuals compared to non-MetS individuals in both subcutaneous and visceral depots. In the abdominal compartment, all the genes showed relatively higher expression in visceral depot as compared to their subcutaneous counterpart, and this difference narrowed with increasing severity of MetS. Their expression level shows an inverse correlation with T2D, MetS, and HOMA-IR and with other T2D-related intermediate traits. Results also demonstrated that individualization of MetS patients could be done based on adipose tissue expression of just 12 genes. Conclusion: Adipose tissue expression of lipodystrophy genes shows an association with MetS and its intermediate phenotypic traits. Mutations of these genes are known to cause congenital lipodystrophy syndromes, whereas their altered expression in adipose tissue contributes to the pathogenesis of IR, T2D, and MetS.


Assuntos
Adipócitos , Diabetes Mellitus Tipo 2 , Resistência à Insulina , Lipodistrofia , Síndrome Metabólica , Humanos , Resistência à Insulina/genética , Síndrome Metabólica/genética , Síndrome Metabólica/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Feminino , Masculino , Adulto , Lipodistrofia/genética , Lipodistrofia/metabolismo , Índia/epidemiologia , Adipócitos/metabolismo , Adipócitos/patologia , Pessoa de Meia-Idade , Povo Asiático/genética , Adulto Jovem , Expressão Gênica
15.
Cells ; 13(19)2024 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-39404417

RESUMO

The ongoing obesity epidemic has raised awareness of the complex physiology of adipose tissue. Abnormal adipocyte differentiation results in the development of systemic metabolic disorders such as insulin resistance and diabetes. The conjugation of NEDD8 (neural precursor cell expressed, developmentally downregulated 8) to target protein, termed neddylation, has been shown to mediate adipogenesis. However, much remains unknown about its role in adipogenesis. Here, we demonstrated that neddylation and its targets, the cullin (CUL) family members, are differentially regulated during mouse and human adipogenesis. Inhibition of neddylation by MLN4924 significantly reduced adipogenesis of 3T3-L1 and human stromal vascular cells. Deletion of NAE1, a subunit of the only NEDD8 E1 enzyme, suppressed neddylation and impaired adipogenesis. Neddylation deficiency did not affect mitotic cell expansion. Instead, it disrupted CREB/CEBPß/PPARγ signaling, essential for adipogenesis. Interestingly, among the neddylation-targeted CUL family members, deletion of CUL3, but not CUL1, CUL2, or CUL4A, largely replicated the adipogenic defects observed with neddylation deficiency. A PPARγ agonist minimally rescued the adipogenic defects caused by the deletion of NAE1 and CUL3. In conclusion, our study demonstrates that neddylation and its targeted CUL3 are crucial for adipogenesis. These findings provide potential targets for therapeutic intervention in obesity and metabolic disorders.


Assuntos
Células 3T3-L1 , Adipócitos , Adipogenia , Diferenciação Celular , Proteínas Culina , Proteína NEDD8 , Proteínas Culina/metabolismo , Adipócitos/metabolismo , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Animais , Humanos , Camundongos , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Proteína NEDD8/metabolismo , Proteína NEDD8/genética , Diferenciação Celular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , PPAR gama/metabolismo , Ciclopentanos/farmacologia , Pirimidinas/farmacologia , Enzimas Ativadoras de Ubiquitina
16.
Pathologie (Heidelb) ; 45(6): 397-403, 2024 Nov.
Artigo em Alemão | MEDLINE | ID: mdl-39365350

RESUMO

A stroma a­reactive invasion front area (SARIFA) is a new prognostic biomarker in carcinomas. Essentially, SARIFA describes the occurrence of direct contact between at least five tumor cells and adipocytes. This phenomenon is extremely easy and quick to identify, shows an extremely low interobserver variability, and does not require any additional staining as it can be identified on standard HE sections. The prognostic efficiency has now been demonstrated in gastric, colorectal, pancreatic, and prostate carcinoma.


Assuntos
Biomarcadores Tumorais , Humanos , Biomarcadores Tumorais/metabolismo , Prognóstico , Adipócitos/patologia , Adipócitos/metabolismo , Invasividade Neoplásica , Masculino , Neoplasias/diagnóstico
17.
Annu Rev Cell Dev Biol ; 40(1): 143-168, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39356808

RESUMO

Lipid droplets (LDs) are dynamic storage organelles with central roles in lipid and energy metabolism. They consist of a core of neutral lipids, such as triacylglycerol, which is surrounded by a monolayer of phospholipids and specialized surface proteins. The surface composition determines many of the LD properties, such as size, subcellular distribution, and interaction with partner organelles. Considering the diverse energetic and metabolic demands of various cell types, it is not surprising that LDs are highly heterogeneous within and between cell types. Despite their diversity, all LDs share a common biogenesis mechanism. However, adipocytes have evolved specific adaptations of these basic mechanisms, enabling the regulation of lipid and energy metabolism at both the cellular and organismal levels. Here, we discuss recent advances in the understanding of both the general mechanisms of LD biogenesis and the adipocyte-specific adaptations controlling these fascinating organelles.


Assuntos
Adipócitos , Gotículas Lipídicas , Gotículas Lipídicas/metabolismo , Humanos , Animais , Adipócitos/metabolismo , Metabolismo dos Lipídeos , Metabolismo Energético
18.
Commun Biol ; 7(1): 1300, 2024 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-39390075

RESUMO

The crosstalk between adipose tissue and the liver is finely controlled to maintain metabolic health. Yet, how adipose tissue controls toxic free fatty acid overflow into the liver remains incompletely understood. Here, we show that adipocyte activating transcription factor 3 (ATF3) was induced in human or mouse obesity. Adipocyte Atf3-/- (Atf3Adi-/-) mice developed obesity, glucose intolerance, and metabolic dysfunction-associated steatohepatitis (MASH) in chow diet, high-fat diet, or Western diet-fed mice. Blocking fatty acid flux by inhibiting hepatocyte CD36, but not the restoration of hepatic AMPK signaling, prevented the aggravation of MASH in Atf3Adi-/- mice. Further studies show that the loss of adipocyte ATF3 increased lipolysis via inducing adipose triglyceride lipase, which in turn induced lipogenesis and inflammation in hepatocytes. Moreover, Atf3Adi-/- mice had reduced energy expenditure and increased adipose lipogenesis and inflammation. Our data demonstrate that adipocyte ATF3 is a gatekeeper in counteracting MASH development under physiological and pathological conditions.


Assuntos
Fator 3 Ativador da Transcrição , Tecido Adiposo , Lipólise , Camundongos Knockout , Animais , Fator 3 Ativador da Transcrição/metabolismo , Fator 3 Ativador da Transcrição/genética , Camundongos , Tecido Adiposo/metabolismo , Humanos , Masculino , Fígado Gorduroso/metabolismo , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Obesidade/metabolismo , Obesidade/genética , Camundongos Endogâmicos C57BL , Adipócitos/metabolismo , Dieta Hiperlipídica/efeitos adversos , Lipogênese/genética , Metabolismo Energético , Feminino
19.
Nat Commun ; 15(1): 8533, 2024 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-39358382

RESUMO

White adipose tissue (WAT) is essential for lipid storage and systemic energy homeostasis. Understanding adipocyte formation and stability is key to developing therapies for obesity and metabolic disorders. Through a high-throughput cDNA screen, we identified PATZ1, a POZ/BTB and AT-Hook Containing Zinc Finger 1 protein, as an important adipogenic transcription factor. PATZ1 is expressed in human and mouse adipocyte precursor cells (APCs) and adipocytes. In cellular models, PATZ1 promotes adipogenesis via protein-protein interactions and DNA binding. PATZ1 ablation in mouse adipocytes and APCs leads to a reduced APC pool, decreased fat mass, and hypertrophied adipocytes. ChIP-Seq and RNA-seq analyses show that PATZ1 supports adipogenesis by interacting with transcriptional machinery at the promoter regions of key early adipogenic factors. Mass-spec results show that PATZ1 associates with GTF2I, with GTF2I modulating PATZ1's function during differentiation. These findings underscore PATZ1's regulatory role in adipocyte differentiation and adiposity, offering insights into adipose tissue development.


Assuntos
Adipócitos , Adipogenia , Regiões Promotoras Genéticas , Fatores de Transcrição , Animais , Feminino , Humanos , Masculino , Camundongos , Células 3T3-L1 , Adipócitos/metabolismo , Adipócitos/citologia , Adipogenia/genética , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/citologia , Diferenciação Celular/genética , Regulação da Expressão Gênica , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética
20.
PLoS One ; 19(10): e0311751, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39401200

RESUMO

Adipose tissue is distributed in diverse locations throughout the human body. Not much is known about the extent to which anatomically distinct adipose depots are functionally distinct, specialized organs, nor whether depot-specific characteristics result from intrinsic developmental programs, as opposed to reversible physiological responses to differences in tissue microenvironment. We used DNA microarrays to compare mRNA expression patterns of isolated human adipocytes and cultured adipose stem cells, before and after ex vivo adipocyte differentiation, from seven anatomically diverse adipose tissue depots. Adipocytes from different depots display distinct gene expression programs, which are most closely shared with anatomically related depots. mRNAs whose expression differs between anatomically diverse groups of depots (e.g., subcutaneous vs. internal) suggest important functional specializations. These depot-specific differences in gene expression were recapitulated when adipocyte progenitor cells from each site were differentiated ex vivo, suggesting that progenitor cells from specific anatomic sites are deterministically programmed to differentiate into depot-specific adipocytes. Many developmental transcription factors show striking depot-specific patterns of expression, suggesting that adipocytes in each anatomic depot are programmed during early development in concert with anatomically related tissues and organs. Our results support the hypothesis that adipocytes from different depots are functionally distinct and that their depot-specific specialization reflects distinct developmental programs.


Assuntos
Adipócitos , RNA Mensageiro , Humanos , Adipócitos/metabolismo , Adipócitos/citologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Feminino , Diferenciação Celular/genética , Tecido Adiposo/metabolismo , Tecido Adiposo/citologia , Masculino , Adulto , Células-Tronco/metabolismo , Células-Tronco/citologia , Pessoa de Meia-Idade , Perfilação da Expressão Gênica , Células Cultivadas , Análise de Sequência com Séries de Oligonucleotídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...