Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Metab ; 4(6): 775-790, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35760872

RESUMO

Obesity induces chronic inflammation resulting in insulin resistance and metabolic disorders. Cold exposure can improve insulin sensitivity in humans and rodents, but the mechanisms have not been fully elucidated. Here, we find that cold resolves obesity-induced inflammation and insulin resistance and improves glucose tolerance in diet-induced obese mice. The beneficial effects of cold exposure on improving obesity-induced inflammation and insulin resistance depend on brown adipose tissue (BAT) and liver. Using targeted liquid chromatography with tandem mass spectrometry, we discovered that cold and ß3-adrenergic stimulation promote BAT to produce maresin 2 (MaR2), a member of the specialized pro-resolving mediators of bioactive lipids that play a role in the resolution of inflammation. Notably, MaR2 reduces inflammation in obesity in part by targeting macrophages in the liver. Thus, BAT-derived MaR2 could contribute to the beneficial effects of BAT activation in resolving obesity-induced inflammation and may inform therapeutic approaches to combat obesity and its complications.


Assuntos
Tecido Adiposo Marrom , Resistência à Insulina , Tecido Adiposo Marrom/metabolismo , Animais , Ácidos Docosa-Hexaenoicos , Inflamação/metabolismo , Camundongos , Obesidade/metabolismo
2.
Nat Metab ; 3(4): 485-495, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33846638

RESUMO

Brown adipose tissue (BAT) and beige fat function in energy expenditure in part due to their role in thermoregulation, making these tissues attractive targets for treating obesity and metabolic disorders. While prolonged cold exposure promotes de novo recruitment of brown adipocytes, the exact sources of cold-induced thermogenic adipocytes are not completely understood. Here, we identify transient receptor potential cation channel subfamily V member 1 (Trpv1)+ vascular smooth muscle (VSM) cells as previously unidentified thermogenic adipocyte progenitors. Single-cell RNA sequencing analysis of interscapular brown adipose depots reveals, in addition to the previously known platelet-derived growth factor receptor (Pdgfr)α-expressing mesenchymal progenitors, a population of VSM-derived adipocyte progenitor cells (VSM-APC) expressing the temperature-sensitive cation channel Trpv1. We demonstrate that cold exposure induces the proliferation of Trpv1+ VSM-APCs and enahnces their differentiation to highly thermogenic adipocytes. Together, these findings illustrate the landscape of the thermogenic adipose niche at single-cell resolution and identify a new cellular origin for the development of brown and beige adipocytes.


Assuntos
Adipócitos/fisiologia , Temperatura Baixa , Células-Tronco Hematopoéticas/fisiologia , Músculo Liso Vascular/fisiologia , Canais de Cátion TRPV/fisiologia , Termogênese/fisiologia , Adipócitos Bege/fisiologia , Adipócitos Marrons/fisiologia , Tecido Adiposo Bege/metabolismo , Tecido Adiposo Marrom/fisiologia , Animais , Regulação da Temperatura Corporal/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Humanos , Células-Tronco Mesenquimais , Camundongos , Camundongos Endogâmicos C57BL , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Canais de Cátion TRPV/genética
3.
Sci Transl Med ; 12(558)2020 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-32848096

RESUMO

Brown and brown-like beige/brite adipocytes dissipate energy and have been proposed as therapeutic targets to combat metabolic disorders. However, the therapeutic effects of cell-based therapy in humans remain unclear. Here, we created human brown-like (HUMBLE) cells by engineering human white preadipocytes using CRISPR-Cas9-SAM-gRNA to activate endogenous uncoupling protein 1 expression. Obese mice that received HUMBLE cell transplants showed a sustained improvement in glucose tolerance and insulin sensitivity, as well as increased energy expenditure. Mechanistically, increased arginine/nitric oxide (NO) metabolism in HUMBLE adipocytes promoted the production of NO that was carried by S-nitrosothiols and nitrite in red blood cells to activate endogenous brown fat and improved glucose homeostasis in recipient animals. Together, these data demonstrate the utility of using CRISPR-Cas9 technology to engineer human white adipocytes to display brown fat-like phenotypes and may open up cell-based therapeutic opportunities to combat obesity and diabetes.


Assuntos
Adipócitos Marrons , Síndrome Metabólica , Tecido Adiposo Marrom/metabolismo , Animais , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Dieta Hiperlipídica , Metabolismo Energético , Humanos , Síndrome Metabólica/terapia , Camundongos , Camundongos Obesos , Obesidade/metabolismo , Obesidade/terapia , Termogênese
4.
Nat Commun ; 11(1): 1421, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32184391

RESUMO

Uncoupling protein-1 (UCP1) plays a central role in energy dissipation in brown adipose tissue (BAT). Using high-throughput library screening of secreted peptides, we identify two fibroblast growth factors (FGF), FGF6 and FGF9, as potent inducers of UCP1 expression in adipocytes and preadipocytes. Surprisingly, this occurs through a mechanism independent of adipogenesis and involves FGF receptor-3 (FGFR3), prostaglandin-E2 and interaction between estrogen receptor-related alpha, flightless-1 (FLII) and leucine-rich-repeat-(in FLII)-interacting-protein-1 as a regulatory complex for UCP1 transcription. Physiologically, FGF6/9 expression in adipose is upregulated by exercise and cold in mice, and FGF9/FGFR3 expression in human neck fat is significantly associated with UCP1 expression. Loss of FGF9 impairs BAT thermogenesis. In vivo administration of FGF9 increases UCP1 expression and thermogenic capacity. Thus, FGF6 and FGF9 are adipokines that can regulate UCP1 through a transcriptional network that is dissociated from brown adipogenesis, and act to modulate systemic energy metabolism.


Assuntos
Adipócitos Marrons/metabolismo , Adipogenia , Fator 6 de Crescimento de Fibroblastos/metabolismo , Fator 9 de Crescimento de Fibroblastos/metabolismo , Obesidade/metabolismo , Proteína Desacopladora 1/metabolismo , Adipócitos Marrons/citologia , Tecido Adiposo Marrom/citologia , Tecido Adiposo Marrom/metabolismo , Animais , Fator 6 de Crescimento de Fibroblastos/genética , Fator 9 de Crescimento de Fibroblastos/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/genética , Obesidade/fisiopatologia , Termogênese , Proteína Desacopladora 1/genética
5.
PLoS Biol ; 18(2): e3000630, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32040503

RESUMO

Opsin3 (Opn3) is a transmembrane heptahelical G protein-coupled receptor (GPCR) with the potential to produce a nonvisual photoreceptive effect. Interestingly, anatomical profiling of GPCRs reveals that Opn3 mRNA is highly expressed in adipose tissue. The photosensitive functions of Opn3 in mammals are poorly understood, and whether Opn3 has a role in fat is entirely unknown. In this study, we found that Opn3-knockout (Opn3-KO) mice were prone to diet-induced obesity and insulin resistance. At the cellular level, Opn3-KO brown adipocytes cultured in darkness had decreased glucose uptake and lower nutrient-induced mitochondrial respiration than wild-type (WT) cells. Light exposure promoted mitochondrial activity and glucose uptake in WT adipocytes but not in Opn3-KO cells. Brown adipocytes carrying a defective mutation in Opn3's putative G protein-binding domain also exhibited a reduction in glucose uptake and mitochondrial respiration in darkness. Using RNA-sequencing, we identified several novel light-sensitive and Opn3-dependent molecular signatures in brown adipocytes. Importantly, direct exposure of brown adipose tissue (BAT) to light in living mice significantly enhanced thermogenic capacity of BAT, and this effect was diminished in Opn3-KO animals. These results uncover a previously unrecognized cell-autonomous, light-sensing mechanism in brown adipocytes via Opn3-GPCR signaling that can regulate fuel metabolism and mitochondrial respiration. Our work also provides a molecular basis for developing light-based treatments for obesity and its related metabolic disorders.


Assuntos
Adipócitos Marrons/metabolismo , Metabolismo Energético , Opsinas de Bastonetes/metabolismo , Tecido Adiposo Marrom/inervação , Animais , Dieta Hiperlipídica/efeitos adversos , Regulação da Expressão Gênica , Glucose/metabolismo , Resistência à Insulina , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mutação , Obesidade/genética , Obesidade/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Opsinas de Bastonetes/genética , Transdução de Sinais , Termogênese
6.
Cell Metab ; 30(4): 768-783.e7, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31353262

RESUMO

Distinct oxygenases and their oxylipin products have been shown to participate in thermogenesis by mediating physiological adaptations required to sustain body temperature. Since the role of the lipoxygenase (LOX) family in cold adaptation remains elusive, we aimed to investigate whether, and how, LOX activity is required for cold adaptation and to identify LOX-derived lipid mediators that could serve as putative cold mimetics with therapeutic potential to combat diabetes. By utilizing mass-spectrometry-based lipidomics in mice and humans, we demonstrated that cold and ß3-adrenergic stimulation could promote the biosynthesis and release of 12-LOX metabolites from brown adipose tissue (BAT). Moreover, 12-LOX ablation in mouse brown adipocytes impaired glucose uptake and metabolism, resulting in blunted adaptation to the cold in vivo. The cold-induced 12-LOX product 12-HEPE was found to be a batokine that improves glucose metabolism by promoting glucose uptake into adipocytes and skeletal muscle through activation of an insulin-like intracellular signaling pathway.


Assuntos
Tecido Adiposo Marrom/metabolismo , Araquidonato 12-Lipoxigenase/fisiologia , Resposta ao Choque Frio/fisiologia , Metabolismo Energético/fisiologia , Obesidade/metabolismo , Adipócitos Marrons/metabolismo , Adipócitos Marrons/patologia , Animais , Linhagem Celular , Feminino , Glucose/metabolismo , Humanos , Masculino , Camundongos , Termogênese/fisiologia
7.
Cell Rep ; 24(3): 781-790, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-30021173

RESUMO

Thermogenic fat expends energy during cold for temperature homeostasis, and its activity regulates nutrient metabolism and insulin sensitivity. We measured cold-activated lipid landscapes in circulation and in adipose tissue by MS/MSALL shotgun lipidomics. We created an interactive online viewer to visualize the changes of specific lipid species in response to cold. In adipose tissue, among the approximately 1,600 lipid species profiled, we identified the biosynthetic pathway of the mitochondrial phospholipid cardiolipin as coordinately activated in brown and beige fat by cold in wild-type and transgenic mice with enhanced browning of white fat. Together, these data provide a comprehensive lipid bio-signature of thermogenic fat activation in circulation and tissue and suggest pathways regulated by cold exposure.


Assuntos
Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Cardiolipinas/metabolismo , Temperatura Baixa , Lipídeos/sangue , Termogênese , Animais , Biomarcadores/sangue , Humanos , Metabolômica , Camundongos , Modelos Animais , Fosfatidilgliceróis/metabolismo
9.
Nat Med ; 23(5): 631-637, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28346411

RESUMO

Brown adipose tissue (BAT) and beige adipose tissue combust fuels for heat production in adult humans, and so constitute an appealing target for the treatment of metabolic disorders such as obesity, diabetes and hyperlipidemia. Cold exposure can enhance energy expenditure by activating BAT, and it has been shown to improve nutrient metabolism. These therapies, however, are time consuming and uncomfortable, demonstrating the need for pharmacological interventions. Recently, lipids have been identified that are released from tissues and act locally or systemically to promote insulin sensitivity and glucose tolerance; as a class, these lipids are referred to as 'lipokines'. Because BAT is a specialized metabolic tissue that takes up and burns lipids and is linked to systemic metabolic homeostasis, we hypothesized that there might be thermogenic lipokines that activate BAT in response to cold. Here we show that the lipid 12,13-dihydroxy-9Z-octadecenoic acid (12,13-diHOME) is a stimulator of BAT activity, and that its levels are negatively correlated with body-mass index and insulin sensitivity. Using a global lipidomic analysis, we found that 12,13-diHOME was increased in the circulation of humans and mice exposed to cold. Furthermore, we found that the enzymes that produce 12,13-diHOME were uniquely induced in BAT by cold stimulation. The injection of 12,13-diHOME acutely activated BAT fuel uptake and enhanced cold tolerance, which resulted in decreased levels of serum triglycerides. Mechanistically, 12,13-diHOME increased fatty acid (FA) uptake into brown adipocytes by promoting the translocation of the FA transporters FATP1 and CD36 to the cell membrane. These data suggest that 12,13-diHOME, or a functional analog, could be developed as a treatment for metabolic disorders.


Assuntos
Tecido Adiposo Marrom/metabolismo , Temperatura Baixa , Ácidos Graxos/metabolismo , Resistência à Insulina , Obesidade/metabolismo , Ácidos Oleicos/metabolismo , Termogênese , Tecido Adiposo Marrom/efeitos dos fármacos , Animais , Transporte Biológico/efeitos dos fármacos , Antígenos CD36/efeitos dos fármacos , Antígenos CD36/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Metabolismo Energético/efeitos dos fármacos , Proteínas de Transporte de Ácido Graxo/efeitos dos fármacos , Proteínas de Transporte de Ácido Graxo/metabolismo , Feminino , Fluordesoxiglucose F18 , Humanos , Masculino , Camundongos , Ácidos Oleicos/biossíntese , Ácidos Oleicos/farmacologia , Sobrepeso/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , RNA Mensageiro/metabolismo , Compostos Radiofarmacêuticos , Triglicerídeos/metabolismo
10.
Diabetologia ; 59(8): 1769-77, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27209464

RESUMO

AIMS/HYPOTHESIS: Adipose tissue dysfunction is a prime risk factor for the development of metabolic disease. Bone morphogenetic proteins (BMPs) have previously been implicated in adipocyte formation. Here, we investigate the role of BMP signalling in adipose tissue health and systemic glucose homeostasis. METHODS: We employed the Cre/loxP system to generate mouse models with conditional ablation of BMP receptor 1A in differentiating and mature adipocytes, as well as tissue-resident myeloid cells. Metabolic variables were assessed by glucose and insulin tolerance testing, insulin-stimulated glucose uptake and gene expression analysis. RESULTS: Conditional deletion of Bmpr1a using the aP2 (also known as Fabp4)-Cre strain resulted in a complex phenotype. Knockout mice were clearly resistant to age-related impairment of insulin sensitivity during normal and high-fat-diet feeding and showed significantly improved insulin-stimulated glucose uptake in brown adipose tissue and skeletal muscle. Moreover, knockouts displayed significant reduction of variables of adipose tissue inflammation. Deletion of Bmpr1a in myeloid cells had no impact on insulin sensitivity, while ablation of Bmpr1a in mature adipocytes partially recapitulated the initial phenotype from aP2-Cre driven deletion. Co-cultivation of macrophages with pre-adipocytes lacking Bmpr1a markedly reduced expression of proinflammatory genes. CONCLUSIONS/INTERPRETATION: Our findings show that altered BMP signalling in adipose tissue affects the tissue's metabolic properties and systemic insulin resistance by altering the pattern of immune cell infiltration. The phenotype is due to ablation of Bmpr1a specifically in pre-adipocytes and maturing adipocytes rather than an immune cell-autonomous effect. Mechanistically, we provide evidence for a BMP-mediated direct crosstalk between pre-adipocytes and macrophages.


Assuntos
Tecido Adiposo/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Resistência à Insulina/fisiologia , Adipócitos/metabolismo , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos não Esterificados/sangue , Glucose/metabolismo , Insulina/sangue , Resistência à Insulina/genética , Interleucina-6/sangue , Camundongos , Camundongos Knockout , Fator de Necrose Tumoral alfa/sangue
11.
EMBO Rep ; 16(10): 1378-93, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26303948

RESUMO

Brown adipose tissue (BAT) dissipates chemical energy as heat and can counteract obesity. MicroRNAs are emerging as key regulators in development and disease. Combining microRNA and mRNA microarray profiling followed by bioinformatic analyses, we identified miR-455 as a new regulator of brown adipogenesis. miR-455 exhibits a BAT-specific expression pattern and is induced by cold and the browning inducer BMP7. In vitro gain- and loss-of-function studies show that miR-455 regulates brown adipocyte differentiation and thermogenesis. Adipose-specific miR-455 transgenic mice display marked browning of subcutaneous white fat upon cold exposure. miR-455 activates AMPKα1 by targeting HIF1an, and AMPK promotes the brown adipogenic program and mitochondrial biogenesis. Concomitantly, miR-455 also targets the adipogenic suppressors Runx1t1 and Necdin, initiating adipogenic differentiation. Taken together, the data reveal a novel microRNA-regulated signaling network that controls brown adipogenesis and may be a potential therapeutic target for human metabolic disorders.


Assuntos
Adipócitos Marrons/metabolismo , Adipogenia/genética , MicroRNAs/genética , Transdução de Sinais , Proteínas Quinases Ativadas por AMP/metabolismo , Tecido Adiposo Branco , Animais , Diferenciação Celular/genética , Células Cultivadas , Temperatura Baixa , Humanos , Camundongos , Camundongos Transgênicos , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Biogênese de Organelas , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Proteínas Repressoras/metabolismo , Termogênese/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
12.
Nat Med ; 21(7): 760-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26076036

RESUMO

Targeting brown adipose tissue (BAT) content or activity has therapeutic potential for treating obesity and the metabolic syndrome by increasing energy expenditure. However, both inter- and intra-individual differences contribute to heterogeneity in human BAT and potentially to differential thermogenic capacity in human populations. Here we generated clones of brown and white preadipocytes from human neck fat and characterized their adipogenic and thermogenic differentiation. We combined an uncoupling protein 1 (UCP1) reporter system and expression profiling to define novel sets of gene signatures in human preadipocytes that could predict the thermogenic potential of the cells once they were maturated. Knocking out the positive UCP1 regulators, PREX1 and EDNRB, in brown preadipocytes using CRISPR-Cas9 markedly abolished the high level of UCP1 in brown adipocytes differentiated from the preadipocytes. Finally, we were able to prospectively isolate adipose progenitors with great thermogenic potential using the cell surface marker CD29. These data provide new insights into the cellular heterogeneity in human fat and offer potential biomarkers for identifying thermogenically competent preadipocytes.


Assuntos
Adipócitos Marrons/metabolismo , Adipócitos Brancos/metabolismo , Biomarcadores/metabolismo , Perfilação da Expressão Gênica , Termogênese/genética , Adipócitos Marrons/citologia , Adipócitos Brancos/citologia , Diferenciação Celular , Linhagem Celular Transformada , Membrana Celular/metabolismo , Células Clonais , Genes Reporter , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Integrina beta1/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor de Endotelina B , Receptores de Endotelina/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteína Desacopladora 1
13.
Cell ; 156(3): 440-55, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24485453

RESUMO

Lung stem cells are instructed to produce lineage-specific progeny through unknown factors in their microenvironment. We used clonal 3D cocultures of endothelial cells and distal lung stem cells, bronchioalveolar stem cells (BASCs), to probe the instructive mechanisms. Single BASCs had bronchiolar and alveolar differentiation potential in lung endothelial cell cocultures. Gain- and loss-of-function experiments showed that BMP4-Bmpr1a signaling triggers calcineurin/NFATc1-dependent expression of thrombospondin-1 (Tsp1) in lung endothelial cells to drive alveolar lineage-specific BASC differentiation. Tsp1 null mice exhibited defective alveolar injury repair, confirming a crucial role for the BMP4-NFATc1-TSP1 axis in lung epithelial differentiation and regeneration in vivo. Discovery of this pathway points to methods to direct the derivation of specific lung epithelial lineages from multipotent cells. These findings elucidate a pathway that may be a critical target in lung diseases and provide tools to understand the mechanisms of respiratory diseases at the single-cell level.


Assuntos
Bronquíolos/citologia , Diferenciação Celular , Células Endoteliais/metabolismo , Alvéolos Pulmonares/citologia , Transdução de Sinais , Células-Tronco/metabolismo , Animais , Proteína Morfogenética Óssea 4/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Bronquíolos/metabolismo , Células Cultivadas , Técnicas de Cocultura , Camundongos , Fatores de Transcrição NFATC/metabolismo , Alvéolos Pulmonares/metabolismo , Células-Tronco/citologia , Trombospondina 1/genética , Trombospondina 1/metabolismo
14.
J Nucl Med ; 54(11): 1896-901, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24071505

RESUMO

UNLABELLED: For brown adipose tissue (BAT) to be effective at consuming calories, its blood flow must increase enough to provide sufficient fuel to sustain energy expenditure and also transfer the heat created to avoid thermal injury. Here we used a combination of human and rodent models to assess changes in BAT blood flow and glucose utilization. METHODS: (99m)Tc-methoxyisobutylisonitrile (MIBI) SPECT (n = 7) and SPECT/CT (n = 74) scans done in adult humans for parathyroid imaging were reviewed for uptake in regions consistent with human BAT. Site-directed biopsies of subcutaneous and deep neck fat were obtained for electron microscopy and gene expression profiling. In mice, tissue perfusion was measured with (99m)Tc-MIBI (n = 16) and glucose uptake with (18)F-FDG (n = 16). Animals were kept fasting overnight, anesthetized with pentobarbital, and given intraperitoneally either the ß3-adrenergic receptor agonist CL-316,243, 1 mg/kg (n = 8), or saline (n = 8) followed by radiotracer injection 5 min later. After 120 min, the mice were imaged using SPECT/CT or PET/CT. Vital signs were recorded over 30 min during the imaging. BAT, white adipose tissue (WAT), muscle, liver, and heart were resected, and tissue uptake of both (99m)Tc-MIBI and (18)F-FDG was quantified by percentage injected dose per gram of tissue and normalized to total body weight. RESULTS: In 5.4% of patients (4/74), (99m)Tc-MIBI SPECT/CT showed increased retention in cervical and supraclavicular fat that displayed multilocular lipid droplets, dense capillary investment, and a high concentration of ovoid mitochondria. Expression levels of the tissue-specific uncoupling protein-1 were 180 times higher in BAT than in subcutaneous WAT (P < 0.001). In mice, BAT tissue perfusion increased by 61% (P < 0.01), with no significant changes in blood flow to WAT, muscle, heart, or liver. CL-316,243 increased glucose uptake in BAT even more, by 440% (P < 0.01). CONCLUSION: Pharmacologic activation of BAT requires increased blood flow to deliver glucose and oxygen for thermogenesis. However, the glucose consumption far exceeds the vascular response. These findings demonstrate that activated BAT increases glucose uptake beyond what might occur by increased blood flow alone and suggest that activated BAT likely uses glucose for nonthermogenic purposes.


Assuntos
Tecido Adiposo Marrom/diagnóstico por imagem , Fluordesoxiglucose F18 , Tomografia por Emissão de Pósitrons , Tecnécio Tc 99m Sestamibi , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X , Tecido Adiposo Marrom/citologia , Tecido Adiposo Marrom/metabolismo , Adulto , Animais , Transporte Biológico , Circulação Sanguínea , Glucose/metabolismo , Humanos , Masculino , Camundongos , Imagem Multimodal , Transcriptoma
15.
Nature ; 495(7441): 379-83, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23485971

RESUMO

Maintenance of body temperature is essential for the survival of homeotherms. Brown adipose tissue (BAT) is a specialized fat tissue that is dedicated to thermoregulation. Owing to its remarkable capacity to dissipate stored energy and its demonstrated presence in adult humans, BAT holds great promise for the treatment of obesity and metabolic syndrome. Rodent data suggest the existence of two types of brown fat cells: constitutive BAT (cBAT), which is of embryonic origin and anatomically located in the interscapular region of mice; and recruitable BAT (rBAT), which resides within white adipose tissue (WAT) and skeletal muscle, and has alternatively been called beige, brite or inducible BAT. Bone morphogenetic proteins (BMPs) regulate the formation and thermogenic activity of BAT. Here we use mouse models to provide evidence for a systemically active regulatory mechanism that controls whole-body BAT activity for thermoregulation and energy homeostasis. Genetic ablation of the type 1A BMP receptor (Bmpr1a) in brown adipogenic progenitor cells leads to a severe paucity of cBAT. This in turn increases sympathetic input to WAT, thereby promoting the formation of rBAT within white fat depots. This previously unknown compensatory mechanism, aimed at restoring total brown-fat-mediated thermogenic capacity in the body, is sufficient to maintain normal temperature homeostasis and resistance to diet-induced obesity. These data suggest an important physiological cross-talk between constitutive and recruitable brown fat cells. This sophisticated regulatory mechanism of body temperature may participate in the control of energy balance and metabolic disease.


Assuntos
Tecido Adiposo Marrom/citologia , Tecido Adiposo Branco/citologia , Proteínas Morfogenéticas Ósseas/metabolismo , Transdução de Sinais , Tecido Adiposo Marrom/inervação , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Temperatura Corporal , Regulação da Temperatura Corporal , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Metabolismo Energético , Camundongos , Células-Tronco/citologia
16.
Antioxid Redox Signal ; 19(3): 243-57, 2013 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22938691

RESUMO

AIMS: Brown adipose tissue dissipates chemical energy in the form of heat and regulates triglyceride and glucose metabolism in the body. Factors that regulate fatty acid uptake and oxidation in brown adipocytes have not yet been fully elucidated. Bone morphogenetic protein 7 (BMP7) is a growth factor capable of inducing brown fat mitochondrial biogenesis during differentiation from adipocyte progenitors. Administration of BMP7 to mice also results in increased energy expenditure. To determine if BMP7 is able to affect the mitochondrial activity of mature brown adipocytes, independent of the differentiation process, we delivered BMP7 to mature brown adipocytes and measured mitochondrial activity. RESULTS: We found that BMP7 increased mitochondrial activity, including fatty acid oxidation and citrate synthase activity, without increasing the mitochondrial number. This was accompanied by an increase in fatty acid uptake and increased protein expression of CPT1 and CD36, which import fatty acids into the mitochondria and the cell, respectively. Importantly, inhibition of either CPT1 or CD36 resulted in a blunting of the mitochondrial activity of BMP7-treated cells. INNOVATION: These findings uncover a novel pathway regulating mitochondrial activities in mature brown adipocytes by BMP7-mediated fatty acid uptake and oxidation. CONCLUSION: In conclusion, BMP7 increases mitochondrial activity in mature brown adipocytes via increased fatty acid uptake and oxidation, a process that requires the fatty acid transporters CPT1 and CD36.


Assuntos
Adipócitos Marrons/metabolismo , Proteína Morfogenética Óssea 7/metabolismo , Antígenos CD36/metabolismo , Carnitina O-Palmitoiltransferase/metabolismo , Ácidos Graxos/metabolismo , Mitocôndrias/metabolismo , Fator 2 Ativador da Transcrição/metabolismo , Trifosfato de Adenosina/metabolismo , Tecido Adiposo Marrom/metabolismo , Animais , Transporte Biológico , Temperatura Corporal , Proteína Morfogenética Óssea 7/genética , Catálise , Respiração Celular , Metabolismo Energético , Expressão Gênica , Camundongos , Modelos Biológicos , Oxirredução , Ácido Pirúvico/metabolismo , Proteínas Smad Reguladas por Receptor/metabolismo , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
FASEB J ; 26(5): 2187-96, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22331196

RESUMO

Body weight is regulated by coordinating energy intake and energy expenditure. Transforming growth factor ß (TGFß)/bone morphogenetic protein (BMP) signaling has been shown to regulate energy balance in lower organisms, but whether a similar pathway exists in mammals is unknown. We have previously demonstrated that BMP7 can regulate brown adipogenesis and energy expenditure. In the current study, we have uncovered a novel role for BMP7 in appetite regulation. Systemic treatment of diet-induced obese mice with BMP7 resulted in increased energy expenditure and decreased food intake, leading to a significant reduction in body weight and improvement of metabolic syndrome. Similar degrees of weight loss with reduced appetite were also observed in BMP7-treated ob/ob mice, suggesting a leptin-independent mechanism utilized by BMP7. Intracerebroventricular administration of BMP7 to mice led to an acute decrease in food intake, which was mediated, at least in part, by a central rapamycin-sensitive mTOR-p70S6 kinase pathway. Together, these results underscore the importance of BMP7 in regulating both food intake and energy expenditure, and suggest new therapeutic approaches for obesity and its comorbidities.


Assuntos
Apetite , Proteína Morfogenética Óssea 7/fisiologia , Obesidade/fisiopatologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Proteína Morfogenética Óssea 7/administração & dosagem , Células Cultivadas , Hibridização In Situ , Injeções Intraventriculares , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Endocrinology ; 152(10): 3680-9, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21862615

RESUMO

Brown adipose tissue plays an important role in obesity, insulin resistance, and diabetes. We have previously shown that the transition from brown preadipocytes to mature adipocytes is mediated in part by insulin receptor substrate (IRS)-1 and the cell cycle regulator protein necdin. In this study, we used pharmacological inhibitors and adenoviral dominant negative constructs to demonstrate that this transition involves IRS-1 activation of Ras and ERK1/2, resulting in phosphorylation of cAMP response element-binding protein (CREB) and suppression of necdin expression. This signaling did not include an elevation of intracellular calcium. A constitutively active form of CREB expressed in IRS-1 knockout cells decreased necdin promoter activity, necdin mRNA, and necdin protein levels, leading to a partial restoration of differentiation. By contrast, forkhead box protein (Fox)O1, which is regulated by the phosphoinositide 3 kinase-Akt pathway, increased necdin promoter activity. Based on reporter gene assays using truncations of the necdin promoter and chromatin immunoprecipitation studies, we demonstrated that CREB and FoxO1 are recruited to the necdin promoter, likely interacting with specific consensus sequences in the proximal region. Based on these results, we propose that insulin/IGF-I act through IRS-1 phosphorylation to stimulate differentiation of brown preadipocytes via two complementary pathways: 1) the Ras-ERK1/2 pathway to activate CREB and 2) the phosphoinositide 3 kinase-Akt pathway to deactivate FoxO1. These two pathways combine to decrease necdin levels and permit the clonal expansion and coordinated gene expression necessary to complete brown adipocyte differentiation.


Assuntos
Adipócitos Marrons/citologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Fatores de Transcrição Forkhead/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Insulina/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/fisiologia , Adipogenia , Animais , Diferenciação Celular , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína Forkhead Box O1 , Proteínas Substratos do Receptor de Insulina/fisiologia , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Fosforilação , Regiões Promotoras Genéticas , Transdução de Sinais
19.
Proc Natl Acad Sci U S A ; 108(1): 143-8, 2011 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-21173238

RESUMO

Brown fat is specialized for energy expenditure and has therefore been proposed to function as a defense against obesity. Despite recent advances in delineating the transcriptional regulation of brown adipocyte differentiation, cellular lineage specification and developmental cues specifying brown-fat cell fate remain poorly understood. In this study, we identify and isolate a subpopulation of adipogenic progenitors (Sca-1(+)/CD45(-)/Mac1(-); referred to as Sca-1(+) progenitor cells, ScaPCs) residing in murine brown fat, white fat, and skeletal muscle. ScaPCs derived from different tissues possess unique molecular expression signatures and adipogenic capacities. Importantly, although the ScaPCs from interscapular brown adipose tissue (BAT) are constitutively committed brown-fat progenitors, Sca-1(+) cells from skeletal muscle and subcutaneous white fat are highly inducible to differentiate into brown-like adipocytes upon stimulation with bone morphogenetic protein 7 (BMP7). Consistent with these findings, human preadipocytes isolated from subcutaneous white fat also exhibit the greatest inducible capacity to become brown adipocytes compared with cells isolated from mesenteric or omental white fat. When muscle-resident ScaPCs are re-engrafted into skeletal muscle of syngeneic mice, BMP7-treated ScaPCs efficiently develop into adipose tissue with brown fat-specific characteristics. Importantly, ScaPCs from obesity-resistant mice exhibit markedly higher thermogenic capacity compared with cells isolated from obesity-prone mice. These data establish the molecular characteristics of tissue-resident adipose progenitors and demonstrate a dynamic interplay between these progenitors and inductive signals that act in concert to specify brown adipocyte development.


Assuntos
Adipócitos Marrons/fisiologia , Tecido Adiposo Branco/citologia , Antígenos Ly/metabolismo , Diferenciação Celular/fisiologia , Proteínas de Membrana/metabolismo , Músculo Esquelético/citologia , Células-Tronco/fisiologia , Adipócitos Marrons/citologia , Animais , Western Blotting , Proteína Morfogenética Óssea 7/metabolismo , Citometria de Fluxo , Imunofluorescência , Humanos , Imuno-Histoquímica , Camundongos , Termogênese/fisiologia
20.
Mol Cell Biol ; 30(17): 4224-33, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20584981

RESUMO

Both insulin and bone morphogenetic protein (BMP) signaling systems are important for adipocyte differentiation. Analysis of gene expression in BMP7-treated fibroblasts revealed a coordinated change in insulin signaling components by BMP7. To further investigate the cross talk between insulin and BMP signaling systems in brown adipogenesis, we examined the effect of BMP7 in insulin receptor substrate 1 (IRS-1)-deficient brown preadipocytes, which exhibit a severe defect in differentiation. Treatment of these cells with BMP7 for 3 days prior to adipogenic induction restored differentiation and expression of brown adipogenic markers. The high level of adipogenic inhibitor preadipocyte factor 1 (Pref-1) in IRS-1-null cells was markedly reduced by 3 days of BMP7 treatment, and analysis of the 1.3-kb pref-1 promoter revealed 9 putative Smad binding elements (SBEs), suggesting that BMP7 could directly suppress Pref-1 expression, thereby allowing the initiation of the adipogenic program. Using a series of sequential deletion mutants of the pref-1 promoter linked to the luciferase gene and chromatin immunoprecipitation, we demonstrate that the promoter-proximal SBE (-192/-184) was critical in mediating BMP7's suppressive effect on pref-1 transcription. Together, these data suggest cross talk between the insulin and BMP signaling systems by which BMP7 can rescue brown adipogenesis in cells with insulin resistance.


Assuntos
Adipogenia , Tecido Adiposo Marrom/citologia , Proteína Morfogenética Óssea 7/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Insulina/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Sequência de Bases , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Ligação ao Cálcio , Células Cultivadas , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Proteínas Substratos do Receptor de Insulina/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Regiões Promotoras Genéticas , Ligação Proteica , Transdução de Sinais , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...