Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Genes Dev ; 31(2): 127-140, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-28202540

RESUMO

The ability to maintain and expand the pool of adipocytes in adults is integral to the regulation of energy balance, tissue/stem cell homeostasis, and disease pathogenesis. For decades, our knowledge of adipocyte precursors has relied on cellular models. The identity of native adipocyte precursors has remained unclear. Recent studies have identified distinct adipocyte precursor populations that are physiologically regulated and contribute to the development, maintenance, and expansion of adipocyte pools in mice. With new tools available, the properties of adipocyte precursors can now be defined, and the regulation and function of adipose plasticity in development and physiology can be explored.


Assuntos
Adipócitos Marrons/citologia , Adipócitos Brancos/citologia , Adipogenia , Animais , Diferenciação Celular , Humanos , Pesquisa/tendências
2.
FASEB J ; 35(4): e21357, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33710685

RESUMO

First-degree relatives (FDRs) of type 2 diabetics (T2D) feature dysfunction of subcutaneous adipose tissue (SAT) long before T2D onset. miRNAs have a role in adipocyte precursor cells (APC) differentiation and in adipocyte identity. Thus, impaired miRNA expression may contribute to SAT dysfunction in FDRs. In the present work, we have explored changes in miRNA expression associated with T2D family history which may affect gene expression in SAT APCs from FDRs. Small RNA-seq was performed in APCs from healthy FDRs and matched controls and omics data were validated by qPCR. Integrative analyses of APC miRNome and transcriptome from FDRs revealed down-regulated hsa-miR-23a-5p, -193a-5p and -193b-5p accompanied by up-regulated Insulin-like Growth Factor 2 (IGF2) gene which proved to be their direct target. The expression changes in these marks were associated with SAT adipocyte hypertrophy in FDRs. APCs from FDRs further demonstrated reduced capability to differentiate into adipocytes. Treatment with IGF2 protein decreased APC adipogenesis, while over-expression of hsa-miR-23a-5p, -193a-5p and -193b-5p enhanced adipogenesis by IGF2 targeting. Indeed, IGF2 increased the Wnt Family Member 10B gene expression in APCs. Down-regulation of the three miRNAs and IGF2 up-regulation was also observed in Peripheral Blood Leukocytes (PBLs) from FDRs. In conclusion, APCs from FDRs feature a specific miRNA/gene profile, which associates with SAT adipocyte hypertrophy and appears to contribute to impaired adipogenesis. PBL detection of this profile may help in identifying adipocyte hypertrophy in individuals at high risk of T2D.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Predisposição Genética para Doença , Fator de Crescimento Insulin-Like II/metabolismo , MicroRNAs/metabolismo , Adipogenia , Clonagem Molecular , Diabetes Mellitus Tipo 2/genética , Família , Regulação da Expressão Gênica , Humanos , Fator de Crescimento Insulin-Like II/genética , MicroRNAs/genética
3.
Am J Physiol Endocrinol Metab ; 320(6): E1148-E1157, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33870712

RESUMO

The cytokine interleukin 4 (IL-4) can increase beige adipogenesis in adult rodents. However, neonatal animals use a distinct adipocyte precursor compartment for adipogenesis as compared with adults. In this study, we address whether IL-4 can induce persistent effects on adipose tissue when administered subcutaneously in the interscapular region during the neonatal period in Sprague-Dawley rats. We injected IL-4 into neonatal male rats during postnatal days 1-6, followed by analysis of adipose tissue and adipocyte precursors at 2 wk and 10 wk of age. Adipocyte precursors were cultured and subjected to differentiation in vitro. We found that a short and transient IL-4 exposure in neonates upregulated uncoupling protein 1 (Ucp1) mRNA expression and decreased fat cell size in subcutaneous white adipose tissue (WAT). Adipocyte precursors from mature rats that had been treated with IL-4 as neonates displayed a decrease in adiponectin (Adipoq) but no change in Ucp1 expression, as compared with controls. Thus, neonatal IL-4 induces acute beige adipogenesis and decreases adipogenic differentiation capacity long term. Overall, these findings indicate that the neonatal period is critical for adipocyte development and may influence the later onset of obesity.NEW & NOTEWORTHY We used neonatal injections in rat to show that IL-4 decreases adipogenesis and increases browning of white fat. In adulthood, adipocyte precursors show persistently decreased adipogenesis but not increased browning. These studies in the neonate are the first, to our knowledge, to show that IL-4 can have long-lasting effects.


Assuntos
Adipogenia/efeitos dos fármacos , Envelhecimento/metabolismo , Interleucina-4/farmacologia , Adipócitos/efeitos dos fármacos , Adipócitos/fisiologia , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/patologia , Envelhecimento/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
4.
Metabolism ; 151: 155716, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37918793

RESUMO

BACKGROUND: The adipocyte hormone adiponectin improves insulin sensitivity and there is an inverse correlation between adiponectin levels and type-2 diabetes risk. Previous research shows that adiponectin remodels the adipose tissue into a more efficient metabolic sink. For instance, mice that overexpress adiponectin show increased capacity for hyperplastic adipose tissue expansion as evident from smaller and metabolically more active white adipocytes. In contrast, the brown adipose tissue (BAT) of these mice looks "whiter" possibly indicating reduced metabolic activity. Here, we aimed to further establish the effect of adiponectin on adipose tissue expansion and adipocyte mitochondrial function as well as to unravel mechanistic aspects in this area. METHODS: Brown and white adipose tissues from adiponectin overexpressing (APN tg) mice and littermate wildtype controls, housed at room and cold temperature, were studied by histological, gene/protein expression and flow cytometry analyses. Metabolic and mitochondrial functions were studied by radiotracers and Seahorse-based technology. In addition, mitochondrial function was assessed in cultured adiponectin deficient adipocytes from APN knockout and heterozygote mice. RESULTS: APN tg BAT displayed increased proliferation prenatally leading to enlarged BAT. Postnatally, APN tg BAT turned whiter than control BAT, confirming previous reports. Furthermore, elevated adiponectin augmented the sympathetic innervation/activation within adipose tissue. APN tg BAT displayed reduced metabolic activity and reduced mitochondrial oxygen consumption rate (OCR). In contrast, APN tg inguinal white adipose tissue (IWAT) displayed enhanced metabolic activity. These metabolic differences between genotypes were apparent also in cultured adipocytes differentiated from BAT and IWAT stroma vascular fraction, and the OCR was reduced in both brown and white APN heterozygote adipocytes. In both APN tg BAT and IWAT, the mesenchymal stem cell-related genes were upregulated along with an increased abundance of Lineage-Sca1+CD34- "beige-like" adipocyte precursor cells. In vitro, the adiponectin receptor agonist Adiporon increased the expression of the proliferation marker Pcna and decreased the expression of Cd34 in Sca1+ mesenchymal stem cells. CONCLUSIONS: We propose that the seemingly opposite effect of adiponectin on BAT and IWAT is mediated by a common mechanism; while reduced adiponectin levels are linked to lower adipocyte OCR, elevated adiponectin levels stimulate expansion of adipocyte precursor cells that produce adipocytes with intrinsically higher metabolic rate than classical white but lower metabolic rate than classical brown adipocytes. Moreover, adiponectin can modify the adipocytes' metabolic activity directly and by enhancing the sympathetic innervation within a fat depot.


Assuntos
Adipócitos Marrons , Adipócitos Brancos , Adiponectina , Termogênese , Animais , Camundongos , Adipócitos Marrons/metabolismo , Adipócitos Brancos/metabolismo , Adiponectina/metabolismo , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Termogênese/genética
5.
Biol Direct ; 18(1): 32, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37322541

RESUMO

Adipose tissue, an organ critical for systemic energy homeostasis, is influenced by type 2 immunity in its development and function. The type 2 cytokine interleukin (IL)-4 induces the proliferation of bipotential adipocyte precursors (APs) in white fat tissue and primes these cells for differentiation into beige adipocytes, which are specialized for thermogenesis. However, the underlying mechanisms have not yet been comprehensively examined. Here, we identified six microRNA (miRNA) genes upregulated upon IL-4 stimulation in APs, miR-322, miR-503, miR-351, miR-542, miR-450a, and miR-450b; these are encoded in the H19X locus of the genome. Their expression is positively regulated by the transcription factor Klf4, whose expression also increases upon IL-4 stimulation. These miRNAs shared a large set of target genes, of which 381 genes were downregulated in mRNA expression upon IL-4 stimulation and enriched in Wnt signaling pathways. Two genes with downregulated expression, Ccnd1 and Fzd6, were repressed by H19X-encoded miRNAs. Additionally, the Wnt signaling activator LiCl downregulated the expression of this group of miRNAs in APs, indicating that Wnt signaling-related genes and these miRNAs form a double-negative feedback regulatory loop. This miRNA/Wnt feedback regulation modulated the elevated proliferation of APs induced by IL-4 stimulation and contributed to priming them for beige adipocyte differentiation. Moreover, the aberrant expression of these miRNAs attenuates the differentiation of APs into beige adipocytes. Collectively, our results suggest that H19X-encoded miRNAs facilitate the transition of APs from proliferation to differentiation in the IL-4-mediated regulation.


Assuntos
MicroRNAs , MicroRNAs/genética , MicroRNAs/metabolismo , Interleucina-4/metabolismo , Diferenciação Celular/genética , Adipócitos/metabolismo , Proliferação de Células
6.
Cell Rep ; 42(2): 112088, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36753417

RESUMO

The hypothalamic pituitary thyroid axis is a major regulator of many differentiation processes, including adipose tissue. However, it remains unclear whether and how thyroid hormone (TH) signaling contributes to preadipocyte commitment and differentiation into mature adipocytes. Here, we show a cell-autonomous effect of TH on the transcriptional regulation of zinc finger protein 423 (Zfp423), an early adipogenic determination factor, in murine adipose depots. Mechanistically, binding of the unliganded TH receptor to a negative TH responsive element within the Zfp423 promoter activates transcriptional activity that is reversed upon TH binding. Zfp423 upregulation is associated with increased GFP+ preadipocyte recruitment in stromal vascular fraction isolated from white fat of hypothyroid Zfp423GFP reporter mice. RNA sequencing identified Zfp423-driven gene programs that are modulated in response to TH during adipogenic differentiation. Collectively, we identified Zfp423 as a key molecule that integrates TH signaling into the regulation of adipose tissue plasticity.


Assuntos
Adipócitos , Proteínas de Ligação a DNA , Animais , Camundongos , Adipócitos/metabolismo , Adipogenia/fisiologia , Tecido Adiposo/metabolismo , Diferenciação Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Obesidade/metabolismo , Hormônios Tireóideos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
Cell Rep ; 42(10): 113196, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37777963

RESUMO

Maintaining healthy adipose tissue is crucial for metabolic health, requiring a deeper understanding of adipocyte development and response to high-calorie diets. This study highlights the importance of TET3 during white adipose tissue (WAT) development and expansion. Selective depletion of Tet3 in adipose precursor cells (APCs) reduces adipogenesis, protects against diet-induced adipose expansion, and enhances whole-body metabolism. Transcriptomic analysis of wild-type and Tet3 knockout (KO) APCs unveiled TET3 target genes, including Pparg and several genes linked to the extracellular matrix, pivotal for adipogenesis and remodeling. DNA methylation profiling and functional studies underscore the importance of DNA demethylation in gene regulation. Remarkably, targeted DNA demethylation at the Pparg promoter restored its transcription. In conclusion, TET3 significantly governs adipogenesis and diet-induced adipose expansion by regulating key target genes in APCs.


Assuntos
Tecido Adiposo , Dioxigenases , Animais , Humanos , Camundongos , Adipócitos/metabolismo , Adipogenia/genética , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/metabolismo , Diferenciação Celular/genética , Dieta , Dioxigenases/metabolismo , Obesidade/genética , Obesidade/metabolismo , PPAR gama/metabolismo
8.
Cell Metab ; 34(5): 783-799.e7, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35447091

RESUMO

Single-cell RNA sequencing (scRNA-seq) has revealed that adult white adipose tissue (WAT) harbors functionally diverse subpopulations of mesenchymal stromal cells that differentially impact tissue plasticity. To date, the molecular basis of this cellular heterogeneity has not been fully defined. Here, we describe a multilayered omics approach to dissect adipose progenitor cell heterogeneity in three dimensions: progenitor subpopulation, sex, and anatomical localization. We applied state-of-the-art mass spectrometry methods to quantify 4,870 proteins in eight different stromal cell populations from perigonadal and inguinal WAT of male and female mice and acquired transcript expression levels of 15,477 genes using RNA-seq. Our data unveil molecular signatures defining sex differences in preadipocyte differentiation and identify regulatory pathways that functionally distinguish adipose progenitor subpopulations. This multilayered omics analysis, freely accessible at http://preadprofiler.net/, provides unprecedented insights into adipose stromal cell heterogeneity and highlights the benefit of complementary proteomics to support findings from scRNA-seq studies.


Assuntos
Adipócitos , Adipogenia , Adipócitos/metabolismo , Tecido Adiposo , Tecido Adiposo Branco/metabolismo , Animais , Diferenciação Celular , Feminino , Masculino , Camundongos , Células-Tronco/metabolismo
9.
Methods Mol Biol ; 2448: 203-215, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35167099

RESUMO

Techniques to trace and isolate brown adipocyte precursor and adipocytes during development and disease are essential to fully understand brown adipose tissue development and function. Here we report several protocols using the R26R-mTmG reporter mice in thermogenic tissues based on confocal microscopy and fluorescence based flow cytometry. These techniques may be useful to understand the influence of genetic or environmental alterations in brown adipocyte precursors and adipocyte biology.


Assuntos
Adipócitos Marrons , Tecido Adiposo Marrom , Adipogenia , Animais , Citometria de Fluxo , Camundongos , Termogênese/genética
10.
Cell Stem Cell ; 28(4): 685-701.e7, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33539723

RESUMO

Adipose precursor cells (APCs) exhibit regional variation in response to obesity, for unclear reasons. Here, we reveal that HIFα-induced PDGFRß signaling within murine white adipose tissue (WAT) PDGFRß+ cells drives inhibitory serine 112 (S112) phosphorylation of PPARγ, the master regulator of adipogenesis. Levels of PPARγ S112 phosphorylation in WAT PDGFRß+ cells are depot dependent, with levels of PPARγ phosphorylation in PDGFRß+ cells inversely correlating with their capacity for adipogenesis upon high-fat-diet feeding. HIFα suppression in PDGFRß+ progenitors promotes subcutaneous and intra-abdominal adipogenesis, healthy WAT remodeling, and improved metabolic health in obesity. These metabolic benefits are mimicked by treatment of obese mice with the PDGFR antagonist Imatinib, which promotes adipocyte hyperplasia and glucose tolerance in a progenitor cell PPARγ-dependent manner. Our studies unveil a mechanism underlying depot-specific responses of APCs to high-fat feeding and highlight the potential for APCs to be targeted pharmacologically to improve metabolic health in obesity.


Assuntos
Adipogenia , Tecido Adiposo , Adipócitos , Tecido Adiposo Branco , Animais , Dieta Hiperlipídica , Camundongos , Camundongos Endogâmicos C57BL , Obesidade
11.
Front Endocrinol (Lausanne) ; 11: 613606, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33679601

RESUMO

Maintenance of adipocyte precursors is critical for regulating metabolism and preventing obesity related disease. These precursors have been immortalized and studied in cellular models as well as-more recently-in animal models. However, little is known about adipocyte precursors from animals of different ages. Most research has focused on adipocyte precursors during obesity. This review goes over the most recent reports of adipocyte precursors during development and in adulthood. Some of these new analyses are due to new techniques such as single cell-RNA sequencing and temporally controlled lineage tracing. With these tools, we have been able to further our understanding of adipocyte precursor lineages and their different regulatory mechanisms. As we learn more about adipocyte precursor plasticity and regulation, we can hope to use this knowledge for future clinical applications.


Assuntos
Adipócitos/metabolismo , Adipogenia/fisiologia , Tecido Adiposo/metabolismo , Obesidade/metabolismo , Adipócitos/patologia , Tecido Adiposo/patologia , Animais , Diferenciação Celular/fisiologia , Humanos , Obesidade/patologia , Análise de Célula Única/métodos
12.
Adipocyte ; 8(1): 98-104, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-30848691

RESUMO

Obesity and associated metabolic complications, including diabetes, cardiovascular and hepatic diseases, and certain types of cancers, create a major socioeconomic burden. Obesity is characterized by excessive expansion of white adipose tissue resulting from increased adipocyte size, and enhanced adipocyte precursor cells proliferation and differentiation into mature adipocytes, a process well-defined as adipogenesis. Efforts to develop therapeutically potent strategies to circumvent obesity are impacted by our limited understanding of molecular mechanisms regulating adipogenesis. In this review, we discuss recently discovered molecular mechanisms restraining adipogenesis. In this perspective, the discoveries of white adipose tissue endogenous adipogenesis-regulatory cells (Aregs) that negatively regulate adipocyte differentiation, platelet-derived growth factor receptor isoform α (PDGFRα) activation and downstream signaling that hinder adipocyte precursors differentiation, and a group of obesity-associated non-coding RNAs (ncRNAs) that regulate adipogenesis open up promising therapeutic avenues to prevent and/or treat obesity.


Assuntos
Adipogenia/fisiologia , Obesidade/metabolismo , Obesidade/prevenção & controle , Adipócitos Brancos/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Diferenciação Celular/fisiologia , Humanos , MicroRNAs/genética , RNA Longo não Codificante/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais
13.
Adipocyte ; 7(4): 273-276, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30153756

RESUMO

The circadian clock is an intricate molecular network that paces a variety of physiological process to ~ 24 hour day/night cycles. Whereas the central circadian clock in the brain is primarily entrained by light signals, peripheral circadian clocks, which are in most cells in the body, receive cues not only from the central pacemaker but also endocrine and other systemic and tissue-specific signals. Prior studies have connected peripheral circadian clocks to metabolism, primarily with studies focused on the robust clock in the liver that responds to feeding/fasting cycles. Adipose tissue is also critical for metabolism and adipocytes have circadian clocks. Yet, the role of the circadian clock in adipocytes is poorly understood. Here we describe our studies that revealed components of the circadian clock in primary adipocyte precursor cells (APCs) in mice. We made the surprising discovery of a particularly prominent role for the circadian gene Period 3 (Per3) in the APC clock. Furthermore, we elucidated that Per3 directly regulates an output pathway of the APC clock to modulate the expression of the Kruppel-like factor 15 (Klf15) gene. Finally, we discovered that this clock-Klf15 pathway regulates adipogenesis in APCs. These finding have important implications for our understanding of adipose tissue biology and metabolism and, we speculate, will generate opportunities to develop novel therapeutic strategies based on the context-specific features of the circadian clock in APCs.


Assuntos
Adipócitos/citologia , Adipogenia , Relógios Circadianos , Proteínas Circadianas Period/metabolismo , Adipócitos/metabolismo , Animais , Células Cultivadas , Ritmo Circadiano , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like , Camundongos , Proteínas Circadianas Period/genética , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
Methods Mol Biol ; 1752: 111-121, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29564767

RESUMO

Flow cytometry and fluorescence-activated cell sorting (FACS) techniques have significantly advanced the characterization of adipocyte precursor cell (APC) populations. They allow immunophenotyping, quantification, and isolation of distinct populations, which is critical for understanding adipose tissue development and homeostasis. Here, we describe the identification and purification of adipocyte precursor cells using flow cytometry and FACS, defined by previously established surface marker profiles. In addition, we describe the mouse models and whole adipose tissue visualization techniques that will enable us to characterize the plasticity and the cellular origin of APCs.


Assuntos
Adipócitos/citologia , Citometria de Fluxo/métodos , Adipogenia/fisiologia , Tecido Adiposo/citologia , Animais , Diferenciação Celular/fisiologia , Separação Celular , Camundongos
15.
Cell Rep ; 21(9): 2367-2375, 2017 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-29186676

RESUMO

The generation of new adipocytes from precursor cells (adipogenesis) has implications for systemic metabolism and is a commonly used model for studying the process of cell differentiation in vitro. Previous studies from us and others suggested that the peripheral circadian clock can influence adipogenesis in vitro, but the mechanisms driving this activity and the relevance for adipogenesis in vivo are unknown. Here we reveal that mouse adipocyte precursor cells (APCs) contain a circadian clock that oscillates in vivo. We expose context-specific features of the clock in APCs: expression of the canonical core clock component Per1 does not significantly oscillate, whereas the lesser-understood paralog Per3 has a prominent rhythm. We discovered that deletion of Per3 promotes adipogenesis in vivo by a clock output pathway in which PER3 and BMAL1 directly regulate Klf15 expression. These findings demonstrate that Per3 has a major role in the APC clock and regulates adipogenesis in vivo.


Assuntos
Adipogenia/fisiologia , Relógios Circadianos/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas Circadianas Period/metabolismo , Fatores de Transcrição/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Adipogenia/genética , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Relógios Circadianos/genética , Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Proteínas de Ligação a DNA/genética , Fatores de Transcrição Kruppel-Like , Camundongos , Proteínas Circadianas Period/genética , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/genética
16.
Adipocyte ; 6(3): 224-233, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28792785

RESUMO

Over the past 2 decades, the incidence of childhood obesity has risen dramatically. This recent rise in childhood obesity is particularly concerning as adults who were obese during childhood develop type II diabetes that is intractable to current forms of treatment compared with individuals who develop obesity in adulthood. While the mechanisms responsible for the exacerbated diabetic phenotype associated with childhood obesity is not clear, it is well known that childhood is an important time period for the establishment of normal white adipose tissue in humans. This association suggests that exposure to obesogenic stimuli during adipose development may have detrimental effects on adipose function and metabolic homeostasis. In this study, we identify the period of development associated with puberty, postnatal days 18-34, as critical for the establishment of normal adipose mass in mice. Exposure of mice to high fat diet only during this time period results in metabolic dysfunction, increased leptin expression, and increased adipocyte size in adulthood in the absence of sustained increased fat mass or body weight. These findings indicate that exposure to obesogenic stimuli during critical developmental periods have prolonged effects on adipose tissue function that may contribute to the exacerbated metabolic dysfunctions associated with childhood obesity.


Assuntos
Tecido Adiposo Branco/metabolismo , Puberdade/fisiologia , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/fisiologia , Adiposidade/fisiologia , Animais , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Feminino , Homeostase/fisiologia , Humanos , Leptina/metabolismo , Masculino , Camundongos , Obesidade/metabolismo , Puberdade/metabolismo
17.
Cell Metab ; 26(3): 493-508.e4, 2017 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-28844880

RESUMO

Type 2 cytokines are important signals triggering biogenesis of thermogenic beige adipocytes in white adipose tissue (WAT) during cold acclimation. However, how cold activates type 2 immunity in WAT remains obscure. Here we show that cold-induced type 2 immune responses and beiging in subcutaneous WAT (scWAT) are abrogated in mice with adipose-selective ablation of FGF21 or its co-receptor ß-Klotho, whereas such impairments are reversed by replenishment with chemokine CCL11. Mechanistically, FGF21 acts on adipocytes in an autocrine manner to promote the expression and secretion of CCL11 via activation of ERK1/2, which drives recruitment of eosinophils into scWAT, leading to increases in accumulation of M2 macrophages, and proliferation and commitment of adipocyte precursors into beige adipocytes. These FGF21-elicited type 2 immune responses and beiging are blocked by CCL11 neutralization. Thus, the adipose-derived FGF21-CCL11 axis triggers cold-induced beiging and thermogenesis by coupling sympathetic nervous system to activation of type 2 immunity in scWAT.


Assuntos
Tecido Adiposo Branco/metabolismo , Quimiocina CCL11/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Imunidade , Sistema Nervoso Simpático/metabolismo , Adaptação Fisiológica/efeitos dos fármacos , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Tecido Adiposo Bege/efeitos dos fármacos , Tecido Adiposo Bege/metabolismo , Tecido Adiposo Branco/efeitos dos fármacos , Animais , Comunicação Autócrina/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Temperatura Baixa , Eosinófilos/efeitos dos fármacos , Eosinófilos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Crescimento de Fibroblastos/deficiência , Glucuronidase/metabolismo , Imunidade/efeitos dos fármacos , Proteínas Klotho , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Sistema Nervoso Simpático/efeitos dos fármacos , Termogênese/efeitos dos fármacos
18.
Nutrients ; 8(3): 178, 2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-27011203

RESUMO

The aim of this work was to determine the effect of a fructose rich diet (FRD) consumed by the pregnant mother on the endocrine-metabolic and in vivo and in vitro adipose tissue (AT) functions of the male offspring in adulthood. At 60 days of age, rats born to FRD-fed mothers (F) showed impaired glucose tolerance after glucose overload and high circulating levels of leptin (LEP). Despite the diminished mass of retroperitoneal AT, this tissue was characterized by enhanced LEP gene expression, and hypertrophic adipocytes secreting in vitro larger amounts of LEP. Analyses of stromal vascular fraction composition by flow cytometry revealed a reduced number of adipocyte precursor cells. Additionally, 60 day-old control (C) and F male rats were subjected to control diet (CC and FC animals) or FRD (CF and FF rats) for three weeks. FF animals were heavier and consumed more calories. Their metabolic-endocrine parameters were aggravated; they developed severe hyperglycemia, hypertriglyceridemia, hyperleptinemia and augmented AT mass with hypertrophic adipocytes. Our study highlights that manipulation of maternal diet induced an offspring phenotype mainly imprinted with a severely unhealthy adipogenic process with undesirable endocrine-metabolic consequences, putting them at high risk for developing a diabetic state.


Assuntos
Tecido Adiposo/metabolismo , Fenômenos Fisiológicos da Nutrição Animal , Carboidratos da Dieta/toxicidade , Frutose/toxicidade , Desnutrição/etiologia , Fenômenos Fisiológicos da Nutrição Materna , Síndrome Metabólica/etiologia , Efeitos Tardios da Exposição Pré-Natal , Tecido Adiposo/fisiopatologia , Adiposidade , Fatores Etários , Animais , Biomarcadores/sangue , Glicemia/metabolismo , Ingestão de Energia , Feminino , Leptina/sangue , Masculino , Desnutrição/sangue , Desnutrição/fisiopatologia , Síndrome Metabólica/sangue , Síndrome Metabólica/fisiopatologia , Fenótipo , Gravidez , Ratos Sprague-Dawley , Fatores Sexuais , Aumento de Peso
19.
Methods Mol Biol ; 1467: 81-91, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27417961

RESUMO

The current global obesity epidemic has triggered increased interest in adipose tissue biology. A major area of attention for many is adipose tissue development. A greater understanding of adipocyte ontogeny could be highly beneficial in answering questions about obesity-associated disease. Recent work has shown that a proportion of mature adipocytes in visceral white adipose tissue are derived from Wt1-expressing adipocyte precursor cells. These adipocyte precursor cells reside within the adipose tissue itself, and are a constituent of the stromal vascular fraction (SVF), along with other, non-adipogenic, cell types. Crucially, heterogeneity exists within the adipocyte precursor population, with only a proportion of cells expressing Wt1. Moreover, it appears that this difference in the precursor cells may influence the mature adipocytes, with Wt1-lineage-positive adipocytes having fewer, larger lipid droplets than the Wt1-lineage negative. Using fluorescence-activated cell sorting, based on specific marker profiles, it is possible to isolate the adipocyte precursor cells from the SVF. Subsequently, this population can be divided into Wt1-expressing and non-expressing fractions, therefore permitting further analysis of the two cell populations, and the mature adipocytes derived from them. In this chapter we outline a method by which adipocyte precursor cells can be isolated, and how, using a specific mouse model, Wt1-expressing and non-expressing cells can be separated.


Assuntos
Adipócitos/citologia , Separação Celular/métodos , Citometria de Fluxo/métodos , Proteínas Repressoras/metabolismo , Adipócitos/metabolismo , Tecido Adiposo Branco/citologia , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Camundongos , Proteínas WT1
20.
Adipocyte ; 4(3): 203-11, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26257993

RESUMO

The production of new adipocytes requires the differentiation of adipocyte precursor (AP) cells residing within the adipose tissue stromal-vascular compartment. The objective was to obtain an immortalized primary adipogenic cell line derived from FACS isolated committed APs using the conditional expression of SV40 T antigen. Adipocyte precursors were isolated from white adipose tissue (WAT) using FACS to remove non-adipogenic cell populations from mice expressing a conditionally regulated SV40 T antigen. APs were maintained by continuous culture and induced to undergo adipogenic differentiation. Adipogenesis, determined by Oil Red O staining, was assessed with each passage and compared to wildtype controls. Adipogenic capability was rapidly lost with increased passage number in committed APs with concurrent reduction in cell proliferation and expression of essential late adipogenic genes, including Pparγ and C/ebpα. Thus, FACS purified committed APs have limited capability to undergo expansion and subsequent adipogenic differentiation in vitro even if they are immortalized with the SV40 T antigen.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA