Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.434
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 182(3): 563-577.e20, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32615086

RESUMO

Adipose tissues dynamically remodel their cellular composition in response to external cues by stimulating beige adipocyte biogenesis; however, the developmental origin and pathways regulating this process remain insufficiently understood owing to adipose tissue heterogeneity. Here, we employed single-cell RNA-seq and identified a unique subset of adipocyte progenitor cells (APCs) that possessed the cell-intrinsic plasticity to give rise to beige fat. This beige APC population is proliferative and marked by cell-surface proteins, including PDGFRα, Sca1, and CD81. Notably, CD81 is not only a beige APC marker but also required for de novo beige fat biogenesis following cold exposure. CD81 forms a complex with αV/ß1 and αV/ß5 integrins and mediates the activation of integrin-FAK signaling in response to irisin. Importantly, CD81 loss causes diet-induced obesity, insulin resistance, and adipose tissue inflammation. These results suggest that CD81 functions as a key sensor of external inputs and controls beige APC proliferation and whole-body energy homeostasis.


Assuntos
Adipogenia/genética , Tecido Adiposo Bege/metabolismo , Metabolismo Energético/genética , Quinase 1 de Adesão Focal/metabolismo , Transdução de Sinais/genética , Células-Tronco/metabolismo , Tetraspanina 28/metabolismo , Adipócitos/metabolismo , Tecido Adiposo Bege/citologia , Tecido Adiposo Bege/crescimento & desenvolvimento , Tecido Adiposo Branco/metabolismo , Adulto , Animais , Ataxina-1/metabolismo , Feminino , Fibronectinas/farmacologia , Quinase 1 de Adesão Focal/genética , Humanos , Inflamação/genética , Inflamação/metabolismo , Resistência à Insulina/genética , Integrinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Obesidade/genética , Obesidade/metabolismo , RNA-Seq , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Análise de Célula Única , Células-Tronco/citologia , Tetraspanina 28/genética
2.
Cell ; 179(6): 1289-1305.e21, 2019 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-31761534

RESUMO

Adult mesenchymal stem cells, including preadipocytes, possess a cellular sensory organelle called the primary cilium. Ciliated preadipocytes abundantly populate perivascular compartments in fat and are activated by a high-fat diet. Here, we sought to understand whether preadipocytes use their cilia to sense and respond to external cues to remodel white adipose tissue. Abolishing preadipocyte cilia in mice severely impairs white adipose tissue expansion. We discover that TULP3-dependent ciliary localization of the omega-3 fatty acid receptor FFAR4/GPR120 promotes adipogenesis. FFAR4 agonists and ω-3 fatty acids, but not saturated fatty acids, trigger mitosis and adipogenesis by rapidly activating cAMP production inside cilia. Ciliary cAMP activates EPAC signaling, CTCF-dependent chromatin remodeling, and transcriptional activation of PPARγ and CEBPα to initiate adipogenesis. We propose that dietary ω-3 fatty acids selectively drive expansion of adipocyte numbers to produce new fat cells and store saturated fatty acids, enabling homeostasis of healthy fat tissue.


Assuntos
Adipogenia , Cílios/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adipogenia/efeitos dos fármacos , Tecido Adiposo Branco/metabolismo , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Fator de Ligação a CCCTC/metabolismo , Cromatina/metabolismo , Cílios/efeitos dos fármacos , AMP Cíclico/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/metabolismo
3.
Immunity ; 57(6): 1345-1359.e5, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38692280

RESUMO

Regulatory T (Treg) cells in epidydimal visceral adipose tissue (eVAT) of lean mice and humans regulate metabolic homeostasis. We found that constitutive or punctual depletion of eVAT-Treg cells reined in the differentiation of stromal adipocyte precursors. Co-culture of these precursors with conditional medium from eVAT-Treg cells limited their differentiation in vitro, suggesting a direct effect. Transcriptional comparison of adipocyte precursors, matured in the presence or absence of the eVAT-Treg-conditioned medium, identified the oncostatin-M (OSM) signaling pathway as a key distinction. Addition of OSM to in vitro cultures blocked the differentiation of adipocyte precursors, while co-addition of anti-OSM antibodies reversed the ability of the eVAT-Treg-conditioned medium to inhibit in vitro adipogenesis. Genetic depletion of OSM (specifically in Treg) cells or of the OSM receptor (specifically on stromal cells) strongly impaired insulin sensitivity and related metabolic indices. Thus, Treg-cell-mediated control of local progenitor cells maintains adipose tissue and metabolic homeostasis, a regulatory axis seemingly conserved in humans.


Assuntos
Adipócitos , Diferenciação Celular , Homeostase , Resistência à Insulina , Linfócitos T Reguladores , Animais , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Humanos , Camundongos , Adipócitos/metabolismo , Diferenciação Celular/imunologia , Oncostatina M/metabolismo , Transdução de Sinais , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/citologia , Gordura Intra-Abdominal/imunologia , Células Estromais/metabolismo , Camundongos Endogâmicos C57BL , Técnicas de Cocultura , Adipogenia , Células Cultivadas , Masculino , Tecido Adiposo/metabolismo , Tecido Adiposo/citologia , Meios de Cultivo Condicionados/farmacologia
4.
Cell ; 175(6): 1575-1590.e22, 2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30415840

RESUMO

During aging, stromal functions are thought to be impaired, but little is known whether this stems from changes of fibroblasts. Using population- and single-cell transcriptomics, as well as long-term lineage tracing, we studied whether murine dermal fibroblasts are altered during physiological aging under different dietary regimes that affect longevity. We show that the identity of old fibroblasts becomes undefined, with the fibroblast states present in young skin no longer clearly demarcated. In addition, old fibroblasts not only reduce the expression of genes involved in the formation of the extracellular matrix, but also gain adipogenic traits, paradoxically becoming more similar to neonatal pro-adipogenic fibroblasts. These alterations are sensitive to systemic metabolic changes: long-term caloric restriction reversibly prevents them, whereas a high-fat diet potentiates them. Our results therefore highlight loss of cell identity and the acquisition of adipogenic traits as a mechanism underlying cellular aging, which is influenced by systemic metabolism.


Assuntos
Adipogenia , Senescência Celular , Fibroblastos/metabolismo , Envelhecimento da Pele , Animais , Restrição Calórica , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Camundongos , Camundongos Transgênicos
5.
Cell ; 170(2): 340-351.e12, 2017 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-28709001

RESUMO

Injured skeletal muscle regenerates, but with age or in muscular dystrophies, muscle is replaced by fat. Upon injury, muscle-resident fibro/adipogenic progenitors (FAPs) proliferated and gave rise to adipocytes. These FAPs dynamically produced primary cilia, structures that transduce intercellular cues such as Hedgehog (Hh) signals. Genetically removing cilia from FAPs inhibited intramuscular adipogenesis, both after injury and in a mouse model of Duchenne muscular dystrophy. Blocking FAP ciliation also enhanced myofiber regeneration after injury and reduced myofiber size decline in the muscular dystrophy model. Hh signaling through FAP cilia regulated the expression of TIMP3, a secreted metalloproteinase inhibitor, that inhibited MMP14 to block adipogenesis. A pharmacological mimetic of TIMP3 blocked the conversion of FAPs into adipocytes, pointing to a strategy to combat fatty degeneration of skeletal muscle. We conclude that ciliary Hh signaling by FAPs orchestrates the regenerative response to skeletal muscle injury.


Assuntos
Adipogenia , Proteínas Hedgehog/metabolismo , Músculo Esquelético/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Adipócitos/metabolismo , Animais , Cílios/metabolismo , Distrofina/genética , Metaloproteinase 14 da Matriz/metabolismo , Camundongos , Desenvolvimento Muscular , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Regeneração , Inibidor Tecidual de Metaloproteinase-3/metabolismo
6.
Genes Dev ; 38(15-16): 772-783, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39266447

RESUMO

The distinct anatomic environment in which adipose tissues arise during organogenesis is a principle determinant of their adult expansion capacity. Metabolic disease results from a deficiency in hyperplastic adipose expansion within the dermal/subcutaneous depot; thus, understanding the embryonic origins of dermal adipose is imperative. Using single-cell transcriptomics throughout murine embryogenesis, we characterized cell populations, including Bcl11b + cells, that regulate the development of dermal white adipose tissue (dWAT). We discovered that BCL11b expression modulates the Wnt signaling microenvironment to enable adipogenic differentiation in the dermal compartment. Subcutaneous and visceral adipose arises from a distinct population of Nefl + cells during embryonic organogenesis, whereas Pi16 + /Dpp4 + fibroadipogenic progenitors support obesity-stimulated hypertrophic expansion in the adult. Together, these results highlight the unique regulatory pathways used by anatomically distinct adipose depots, with important implications for human metabolic disease.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas Repressoras , Animais , Camundongos , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Adipogenia/genética , Tecido Adiposo Branco/embriologia , Tecido Adiposo Branco/metabolismo , Via de Sinalização Wnt/genética , Tecido Adiposo/metabolismo , Tecido Adiposo/embriologia , Diferenciação Celular/genética , Humanos
7.
Nat Rev Mol Cell Biol ; 20(4): 242-258, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30610207

RESUMO

Obesity is characterized by increased adipose tissue mass and has been associated with a strong predisposition towards metabolic diseases and cancer. Thus, it constitutes a public health issue of major proportion. The expansion of adipose depots can be driven either by the increase in adipocyte size (hypertrophy) or by the formation of new adipocytes from precursor differentiation in the process of adipogenesis (hyperplasia). Notably, adipocyte expansion through adipogenesis can offset the negative metabolic effects of obesity, and the mechanisms and regulators of this adaptive process are now emerging. Over the past several years, we have learned a considerable amount about how adipocyte fate is determined and how adipogenesis is regulated by signalling and systemic factors. We have also gained appreciation that the adipogenic niche can influence tissue adipogenic capability. Approaches aimed at increasing adipogenesis over adipocyte hypertrophy can now be explored as a means to treat metabolic diseases.


Assuntos
Adipogenia/fisiologia , Adipócitos/metabolismo , Adipócitos/fisiologia , Animais , Diferenciação Celular/fisiologia , Saúde , Humanos , Obesidade/metabolismo , Obesidade/fisiopatologia , Transdução de Sinais/fisiologia
8.
Genes Dev ; 37(17-18): 781-800, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37798016

RESUMO

Adipose tissue exhibits a remarkable capacity to expand, contract, and remodel in response to changes in physiological and environmental conditions. Here, we describe recent advances in our understanding of how functionally distinct tissue-resident mesenchymal stromal cell subpopulations orchestrate several aspects of physiological and pathophysiological adipose tissue remodeling, with a particular focus on the adaptations that occur in response to changes in energy surplus and environmental temperature. The study of adipose tissue remodeling provides a vehicle to understand the functional diversity of stromal cells and offers a lens through which several generalizable aspects of tissue reorganization can be readily observed.


Assuntos
Adipogenia , Células-Tronco Mesenquimais , Humanos , Tecido Adiposo , Obesidade , Células Estromais
9.
Cell ; 160(1-2): 105-18, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25579684

RESUMO

Adipose tissue is an essential regulator of metabolic homeostasis. In contrast with white adipose tissue, which stores excess energy in the form of triglycerides, brown adipose tissue is thermogenic, dissipating energy as heat via the unique expression of the mitochondrial uncoupling protein UCP1. A subset of UCP1+ adipocytes develops within white adipose tissue in response to physiological stimuli; however, the developmental origin of these "brite" or "beige" adipocytes is unclear. Here, we report the identification of a BMP7-ROCK signaling axis regulating beige adipocyte formation via control of the G-actin-regulated transcriptional coactivator myocardin-related transcription factor A, MRTFA. White adipose tissue from MRTFA(-/-) mice contains more multilocular adipocytes and expresses enhanced levels of brown-selective proteins, including UCP1. MRTFA(-/-) mice also show improved metabolic profiles and protection from diet-induced obesity and insulin resistance. Our study hence unravels a central pathway driving the development of physiologically functional beige adipocytes.


Assuntos
Transativadores/metabolismo , Adipogenia , Animais , Proteína Morfogenética Óssea 7/metabolismo , Dieta , Metabolismo Energético , Resistência à Insulina , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Transativadores/genética , Fator de Crescimento Transformador beta1/metabolismo
10.
Nat Immunol ; 18(6): 654-664, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28414311

RESUMO

In obesity, inflammation of white adipose tissue (AT) is associated with diminished generation of beige adipocytes ('beige adipogenesis'), a thermogenic and energy-dissipating function mediated by beige adipocytes that express the uncoupling protein UCP1. Here we delineated an inflammation-driven inhibitory mechanism of beige adipogenesis in obesity that required direct adhesive interactions between macrophages and adipocytes mediated by the integrin α4 and its counter-receptor VCAM-1, respectively; expression of the latter was upregulated in obesity. This adhesive interaction reciprocally and concomitantly modulated inflammatory activation of macrophages and downregulation of UCP1 expression dependent on the kinase Erk in adipocytes. Genetic or pharmacological inactivation of the integrin α4 in mice resulted in elevated expression of UCP1 and beige adipogenesis of subcutaneous AT in obesity. Our findings, established in both mouse systems and human systems, reveal a self-sustained cycle of inflammation-driven impairment of beige adipogenesis in obesity.


Assuntos
Adipócitos Bege , Adipogenia/imunologia , Tecido Adiposo Branco/imunologia , Diferenciação Celular/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Obesidade/imunologia , Células 3T3-L1 , Adipócitos/imunologia , Adipócitos/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Adesão Celular/imunologia , Dieta Hiperlipídica , Regulação para Baixo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Retroalimentação , Feminino , Técnicas de Silenciamento de Genes , Humanos , Immunoblotting , Integrina alfa4/genética , Macrófagos/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Monócitos/imunologia , Obesidade/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Gordura Subcutânea , Linfócitos T/imunologia , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo , Adulto Jovem
11.
Cell ; 156(1-2): 20-44, 2014 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-24439368

RESUMO

There has been an upsurge of interest in the adipocyte coincident with the onset of the obesity epidemic and the realization that adipose tissue plays a major role in the regulation of metabolic function. The past few years, in particular, have seen significant changes in the way that we classify adipocytes and how we view adipose development and differentiation. We have new perspective on the roles played by adipocytes in a variety of homeostatic processes and on the mechanisms used by adipocytes to communicate with other tissues. Finally, there has been significant progress in understanding how these relationships are altered during metabolic disease and how they might be manipulated to restore metabolic health.


Assuntos
Adipócitos/metabolismo , Metabolismo dos Lipídeos , Obesidade/patologia , Adipogenia , Animais , Humanos , Obesidade/metabolismo
12.
Immunity ; 50(1): 121-136.e5, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30594464

RESUMO

Dermal fibroblasts (dFBs) resist infection by locally differentiating into adipocytes and producing cathelicidin antimicrobial peptide in response to Staphylococcus aureus (S. aureus). Here, we show that neonatal skin was enriched with adipogenic dFBs and immature dermal fat that highly expressed cathelicidin. The pool of adipogenic and antimicrobial dFBs declined after birth, leading to an age-dependent loss of dermal fat and a decrease in adipogenesis and cathelidicin production in response to infection. Transforming growth factor beta (TGF-ß), which acted on uncommitted embryonic and adult dFBs and inhibited their adipogenic and antimicrobial function, was identified as a key upstream regulator of this process. Furthermore, inhibition of the TGF-ß receptor restored the adipogenic and antimicrobial function of dFBs in culture and increased resistance of adult mice to S. aureus infection. These results provide insight into changes that occur in the skin innate immune system between the perinatal and adult periods of life.


Assuntos
Envelhecimento/imunologia , Fibroblastos/fisiologia , Pele/metabolismo , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/fisiologia , Gordura Subcutânea/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Adipócitos/metabolismo , Adipogenia , Animais , Anti-Infecciosos/metabolismo , Peptídeos Catiônicos Antimicrobianos/metabolismo , Células Cultivadas , Embrião de Mamíferos , Humanos , Imunidade Inata , Camundongos , Catelicidinas
13.
Mol Cell ; 79(6): 874-875, 2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32946760

RESUMO

PARP enzymes are increasingly taking on important roles beyond DNA repair. Huang et al. (2020b) report how the NAD+-dependent ADP-ribosylation of histone H2B by PARP-1 in complex with a metabolic enzyme suppresses the phosphorylation of an adjacent residue, impacting adipogenesis.


Assuntos
Histonas , Inibidores de Poli(ADP-Ribose) Polimerases , ADP-Ribosilação , Adipogenia , Epigênese Genética , Humanos , Obesidade , Fosforilação , Poli(ADP-Ribose) Polimerases
14.
Mol Cell ; 79(6): 934-949.e14, 2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32822587

RESUMO

Although ADP-ribosylation of histones by PARP-1 has been linked to genotoxic stress responses, its role in physiological processes and gene expression has remained elusive. We found that NAD+-dependent ADP-ribosylation of histone H2B-Glu35 by small nucleolar RNA (snoRNA)-activated PARP-1 inhibits AMP kinase-mediated phosphorylation of adjacent H2B-Ser36, which is required for the proadipogenic gene expression program. The activity of PARP-1 on H2B requires NMNAT-1, a nuclear NAD+ synthase, which directs PARP-1 catalytic activity to Glu and Asp residues. ADP-ribosylation of Glu35 and the subsequent reduction of H2B-Ser36 phosphorylation inhibits the differentiation of adipocyte precursors in cultured cells. Parp1 knockout in preadipocytes in a mouse lineage-tracing genetic model increases adipogenesis, leading to obesity. Collectively, our results demonstrate a functional interplay between H2B-Glu35 ADP-ribosylation and H2B-Ser36 phosphorylation that controls adipogenesis.


Assuntos
ADP-Ribosilação/genética , Adipogenia/genética , Histonas/genética , Poli(ADP-Ribose) Polimerase-1/genética , Adenosina Difosfato Ribose/genética , Adipócitos/metabolismo , Adipócitos/patologia , Animais , Linhagem Celular , Dano ao DNA/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos , Fosforilação/genética , RNA Nucleolar Pequeno/genética
15.
Annu Rev Biochem ; 81: 715-36, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22463691

RESUMO

Excessive caloric intake without a rise in energy expenditure promotes adipocyte hyperplasia and adiposity. The rise in adipocyte number is triggered by signaling factors that induce conversion of mesenchymal stem cells (MSCs) to preadipocytes that differentiate into adipocytes. MSCs, which are recruited from the vascular stroma of adipose tissue, provide an unlimited supply of adipocyte precursors. Members of the BMP and Wnt families are key mediators of stem cell commitment to produce preadipocytes. Following commitment, exposure of growth-arrested preadipocytes to differentiation inducers [insulin-like growth factor 1 (IGF1), glucocorticoid, and cyclic AMP (cAMP)] triggers DNA replication and reentry into the cell cycle (mitotic clonal expansion). Mitotic clonal expansion involves a transcription factor cascade, followed by the expression of adipocyte genes. Critical to these events are phosphorylations of the transcription factor CCATT enhancer-binding protein ß (C/EBPß) by MAP kinase and GSK3ß to produce a conformational change that gives rise to DNA-binding activity. "Activated" C/EBPß then triggers transcription of peroxisome proliferator-activated receptor-γ (PPARγ) and C/EBPα, which in turn coordinately activate genes whose expression produces the adipocyte phenotype.


Assuntos
Adipócitos/citologia , Adipócitos/metabolismo , Adipogenia , Animais , Humanos , Transdução de Sinais , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo
16.
Nat Rev Mol Cell Biol ; 16(8): 461-72, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26177004

RESUMO

Autophagy is a conserved catabolic process that degrades cytoplasmic constituents and organelles in the lysosome. Starvation-induced protein degradation is a salient feature of autophagy but recent progress has illuminated how autophagy, during both starvation and nutrient-replete conditions, can mobilize diverse cellular energy and nutrient stores such as lipids, carbohydrates and iron. Processes such as lipophagy, glycophagy and ferritinophagy enable cells to salvage key metabolites to sustain and facilitate core anabolic functions. Here, we discuss the established and emerging roles of autophagy in fuelling biosynthetic capacity and in promoting metabolic and nutrient homeostasis.


Assuntos
Autofagia , Metabolismo Energético , Adipogenia , Animais , Metabolismo dos Carboidratos , Humanos , Ferro/metabolismo , Fagossomos/fisiologia , Transporte Proteico , Proteólise
17.
Cell ; 149(4): 871-85, 2012 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-22579288

RESUMO

Thermogenesis in brown adipose tissue (BAT) is fundamental to energy balance and is also relevant for humans. Bone morphogenetic proteins (BMPs) regulate adipogenesis, and, here, we describe a role for BMP8B in the direct regulation of thermogenesis. BMP8B is induced by nutritional and thermogenic factors in mature BAT, increasing the response to noradrenaline through enhanced p38MAPK/CREB signaling and increased lipase activity. Bmp8b(-/-) mice exhibit impaired thermogenesis and reduced metabolic rate, causing weight gain despite hypophagia. BMP8B is also expressed in the hypothalamus, and Bmp8b(-/-) mice display altered neuropeptide levels and reduced phosphorylation of AMP-activated protein kinase (AMPK), indicating an anorexigenic state. Central BMP8B treatment increased sympathetic activation of BAT, dependent on the status of AMPK in key hypothalamic nuclei. Our results indicate that BMP8B is a thermogenic protein that regulates energy balance in partnership with hypothalamic AMPK. BMP8B may offer a mechanism to specifically increase energy dissipation by BAT.


Assuntos
Tecido Adiposo Marrom/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Dieta , Obesidade/metabolismo , Termogênese , Proteínas Quinases Ativadas por AMP/metabolismo , Adipogenia , Animais , Proteínas Morfogenéticas Ósseas/genética , Metabolismo Energético , Feminino , Hipotálamo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Norepinefrina/metabolismo , Ratos , Ratos Sprague-Dawley
18.
Mol Cell ; 76(1): 163-176.e8, 2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31492633

RESUMO

Sensing nutrient availability is essential for appropriate cellular growth, and mTORC1 is a major regulator of this process. Mechanisms causing mTORC1 activation are, however, complex and diverse. We report here an additional important step in the activation of mTORC1, which regulates the efflux of amino acids from lysosomes into the cytoplasm. This process requires DRAM-1, which binds the membrane carrier protein SCAMP3 and the amino acid transporters SLC1A5 and LAT1, directing them to lysosomes and permitting efficient mTORC1 activation. Consequently, we show that loss of DRAM-1 also impacts pathways regulated by mTORC1, including insulin signaling, glycemic balance, and adipocyte differentiation. Interestingly, although DRAM-1 can promote autophagy, this effect on mTORC1 is autophagy independent, and autophagy only becomes important for mTORC1 activation when DRAM-1 is deleted. These findings provide important insights into mTORC1 activation and highlight the importance of DRAM-1 in growth control, metabolic homeostasis, and differentiation.


Assuntos
Aminoácidos/metabolismo , Proteína 7 Relacionada à Autofagia/metabolismo , Metabolismo Energético , Lisossomos/enzimologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas de Membrana/metabolismo , Células 3T3-L1 , Adipócitos/enzimologia , Adipogenia , Sistema ASC de Transporte de Aminoácidos/genética , Sistema ASC de Transporte de Aminoácidos/metabolismo , Sistema y+L de Transporte de Aminoácidos/genética , Sistema y+L de Transporte de Aminoácidos/metabolismo , Animais , Proteína 7 Relacionada à Autofagia/genética , Glicemia/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Ativação Enzimática , Células HEK293 , Células HeLa , Humanos , Insulina/sangue , Transportador 1 de Aminoácidos Neutros Grandes/genética , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Antígenos de Histocompatibilidade Menor/genética , Antígenos de Histocompatibilidade Menor/metabolismo , Transporte Proteico
19.
Proc Natl Acad Sci U S A ; 121(3): e2300096121, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38194457

RESUMO

The prevalence of overweight and obesity continues to rise in the population worldwide. Because it is an important predisposing factor for cancer, cardiovascular diseases, diabetes mellitus, and COVID-19, obesity reduces life expectancy. Adipose tissue (AT), the main fat storage organ with endocrine capacity, plays fundamental roles in systemic metabolism and obesity-related diseases. Dysfunctional AT can induce excess or reduced body fat (lipodystrophy). Dido1 is a marker gene for stemness; gene-targeting experiments compromised several functions ranging from cell division to embryonic stem cell differentiation, both in vivo and in vitro. We report that mutant mice lacking the DIDO N terminus show a lean phenotype. This consists of reduced AT and hypolipidemia, even when mice are fed a high-nutrient diet. DIDO mutation caused hypothermia due to lipoatrophy of white adipose tissue (WAT) and dermal fat thinning. Deep sequencing of the epididymal white fat (Epi WAT) transcriptome supported Dido1 control of the cellular lipid metabolic process. We found that, by controlling the expression of transcription factors such as C/EBPα or PPARγ, Dido1 is necessary for adipocyte differentiation, and that restoring their expression reestablished adipogenesis capacity in Dido1 mutants. Our model differs from other lipodystrophic mice and could constitute a new system for the development of therapeutic intervention in obesity.


Assuntos
Adipogenia , Lipodistrofia , Animais , Camundongos , Adipogenia/genética , Diferenciação Celular , Dieta , Obesidade/genética , Sobrepeso
20.
Proc Natl Acad Sci U S A ; 121(24): e2319301121, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38838011

RESUMO

Alcohol dehydrogenase 1B (ADH1B) is a primate-specific enzyme which, uniquely among the ADH class 1 family, is highly expressed both in adipose tissue and liver. Its expression in adipose tissue is reduced in obesity and increased by insulin stimulation. Interference with ADH1B expression has also been reported to impair adipocyte function. To better understand the role of ADH1B in adipocytes, we used CRISPR/Cas9 to delete ADH1B in human adipose stem cells (ASC). Cells lacking ADH1B failed to differentiate into mature adipocytes manifested by minimal triglyceride accumulation and a marked reduction in expression of established adipocyte markers. As ADH1B is capable of converting retinol to retinoic acid (RA), we conducted rescue experiments. Incubation of ADH1B-deficient preadipocytes with 9-cis-RA, but not with all-transretinol, significantly rescued their ability to accumulate lipids and express markers of adipocyte differentiation. A homozygous missense variant in ADH1B (p.Arg313Cys) was found in a patient with congenital lipodystrophy of unknown cause. This variant significantly impaired the protein's dimerization, enzymatic activity, and its ability to rescue differentiation in ADH1B-deficient ASC. The allele frequency of this variant in the Middle Eastern population suggests that it is unlikely to be a fully penetrant cause of severe lipodystrophy. In conclusion, ADH1B appears to play an unexpected, crucial and cell-autonomous role in human adipocyte differentiation by serving as a necessary source of endogenous retinoic acid.


Assuntos
Adipócitos , Adipogenia , Álcool Desidrogenase , Humanos , Álcool Desidrogenase/metabolismo , Álcool Desidrogenase/genética , Adipogenia/genética , Adipócitos/metabolismo , Adipócitos/citologia , Tretinoína/metabolismo , Diferenciação Celular , Sistemas CRISPR-Cas , Mutação de Sentido Incorreto , Tecido Adiposo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA