Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92.959
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38959890

RESUMEN

Hypothalamic neural circuits regulate instinctive behaviors such as food seeking, the fight/flight response, socialization, and maternal care. Here, we identified microdeletions on chromosome Xq23 disrupting the brain-expressed transient receptor potential (TRP) channel 5 (TRPC5). This family of channels detects sensory stimuli and converts them into electrical signals interpretable by the brain. Male TRPC5 deletion carriers exhibited food seeking, obesity, anxiety, and autism, which were recapitulated in knockin male mice harboring a human loss-of-function TRPC5 mutation. Women carrying TRPC5 deletions had severe postpartum depression. As mothers, female knockin mice exhibited anhedonia and depression-like behavior with impaired care of offspring. Deletion of Trpc5 from oxytocin neurons in the hypothalamic paraventricular nucleus caused obesity in both sexes and postpartum depressive behavior in females, while Trpc5 overexpression in oxytocin neurons in knock-in mice reversed these phenotypes. We demonstrate that TRPC5 plays a pivotal role in mediating innate human behaviors fundamental to survival, including food seeking and maternal care.

2.
Cell ; 186(4): 715-731.e19, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36754048

RESUMEN

Transgenerational epigenetic inheritance in mammals remains a debated subject. Here, we demonstrate that DNA methylation of promoter-associated CpG islands (CGIs) can be transmitted from parents to their offspring in mice. We generated DNA methylation-edited mouse embryonic stem cells (ESCs), in which CGIs of two metabolism-related genes, the Ankyrin repeat domain 26 and the low-density lipoprotein receptor, were specifically methylated and silenced. DNA methylation-edited mice generated by microinjection of the methylated ESCs exhibited abnormal metabolic phenotypes. Acquired methylation of the targeted CGI and the phenotypic traits were maintained and transmitted across multiple generations. The heritable CGI methylation was subjected to reprogramming in parental PGCs and subsequently reestablished in the next generation at post-implantation stages. These observations provide a concrete step toward demonstrating transgenerational epigenetic inheritance in mammals, which may have implications in our understanding of evolutionary biology as well as the etiology, diagnosis, and prevention of non-genetically inherited human diseases.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Ratones , Humanos , Animales , Islas de CpG , Patrón de Herencia , Mamíferos/genética
3.
Cell ; 185(3): 419-446, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35120662

RESUMEN

Adipose tissue, colloquially known as "fat," is an extraordinarily flexible and heterogeneous organ. While historically viewed as a passive site for energy storage, we now appreciate that adipose tissue regulates many aspects of whole-body physiology, including food intake, maintenance of energy levels, insulin sensitivity, body temperature, and immune responses. A crucial property of adipose tissue is its high degree of plasticity. Physiologic stimuli induce dramatic alterations in adipose-tissue metabolism, structure, and phenotype to meet the needs of the organism. Limitations to this plasticity cause diminished or aberrant responses to physiologic cues and drive the progression of cardiometabolic disease along with other pathological consequences of obesity.


Asunto(s)
Adaptación Fisiológica , Tejido Adiposo/fisiología , Enfermedad , Salud , Adipocitos Blancos/metabolismo , Animales , Humanos , Termogénesis
4.
Cell ; 185(6): 949-966.e19, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35247329

RESUMEN

Beige fat plays key roles in the regulation of systemic energy homeostasis; however, detailed mechanisms and safe strategy for its activation remain elusive. In this study, we discovered that local hyperthermia therapy (LHT) targeting beige fat promoted its activation in humans and mice. LHT achieved using a hydrogel-based photothermal therapy activated beige fat, preventing and treating obesity in mice without adverse effects. HSF1 is required for the effects since HSF1 deficiency blunted the metabolic benefits of LHT. HSF1 regulates Hnrnpa2b1 (A2b1) transcription, leading to increased mRNA stability of key metabolic genes. Importantly, analysis of human association studies followed by functional analysis revealed that the HSF1 gain-of-function variant p.P365T is associated with improved metabolic performance in humans and increased A2b1 transcription in mice and cells. Overall, we demonstrate that LHT offers a promising strategy against obesity by inducing beige fat activation via HSF1-A2B1 transcriptional axis.


Asunto(s)
Tejido Adiposo Beige , Tejido Adiposo Blanco , Hipertermia Inducida , Obesidad/terapia , Tejido Adiposo Beige/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo
5.
Cell ; 185(19): 3501-3519.e20, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36041436

RESUMEN

How intestinal microbes regulate metabolic syndrome is incompletely understood. We show that intestinal microbiota protects against development of obesity, metabolic syndrome, and pre-diabetic phenotypes by inducing commensal-specific Th17 cells. High-fat, high-sugar diet promoted metabolic disease by depleting Th17-inducing microbes, and recovery of commensal Th17 cells restored protection. Microbiota-induced Th17 cells afforded protection by regulating lipid absorption across intestinal epithelium in an IL-17-dependent manner. Diet-induced loss of protective Th17 cells was mediated by the presence of sugar. Eliminating sugar from high-fat diets protected mice from obesity and metabolic syndrome in a manner dependent on commensal-specific Th17 cells. Sugar and ILC3 promoted outgrowth of Faecalibaculum rodentium that displaced Th17-inducing microbiota. These results define dietary and microbiota factors posing risk for metabolic syndrome. They also define a microbiota-dependent mechanism for immuno-pathogenicity of dietary sugar and highlight an elaborate interaction between diet, microbiota, and intestinal immunity in regulation of metabolic disorders.


Asunto(s)
Síndrome Metabólico , Microbiota , Animales , Dieta Alta en Grasa , Azúcares de la Dieta , Interleucina-17 , Mucosa Intestinal , Lípidos , Ratones , Ratones Endogámicos C57BL , Obesidad , Células Th17
6.
Cell ; 183(7): 1848-1866.e26, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33301708

RESUMEN

Obesity is a major cancer risk factor, but how differences in systemic metabolism change the tumor microenvironment (TME) and impact anti-tumor immunity is not understood. Here, we demonstrate that high-fat diet (HFD)-induced obesity impairs CD8+ T cell function in the murine TME, accelerating tumor growth. We generate a single-cell resolution atlas of cellular metabolism in the TME, detailing how it changes with diet-induced obesity. We find that tumor and CD8+ T cells display distinct metabolic adaptations to obesity. Tumor cells increase fat uptake with HFD, whereas tumor-infiltrating CD8+ T cells do not. These differential adaptations lead to altered fatty acid partitioning in HFD tumors, impairing CD8+ T cell infiltration and function. Blocking metabolic reprogramming by tumor cells in obese mice improves anti-tumor immunity. Analysis of human cancers reveals similar transcriptional changes in CD8+ T cell markers, suggesting interventions that exploit metabolism to improve cancer immunotherapy.


Asunto(s)
Inmunidad , Neoplasias/inmunología , Neoplasias/metabolismo , Obesidad/metabolismo , Microambiente Tumoral , Adiposidad , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Proliferación Celular , Dieta Alta en Grasa , Ácidos Grasos/metabolismo , Células HEK293 , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Cinética , Linfocitos Infiltrantes de Tumor , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Análisis de Componente Principal , Procolágeno-Prolina Dioxigenasa/metabolismo , Proteómica
7.
Cell ; 181(4): 832-847.e18, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32304665

RESUMEN

Obesity is a major modifiable risk factor for pancreatic ductal adenocarcinoma (PDAC), yet how and when obesity contributes to PDAC progression is not well understood. Leveraging an autochthonous mouse model, we demonstrate a causal and reversible role for obesity in early PDAC progression, showing that obesity markedly enhances tumorigenesis, while genetic or dietary induction of weight loss intercepts cancer development. Molecular analyses of human and murine samples define microenvironmental consequences of obesity that foster tumorigenesis rather than new driver gene mutations, including significant pancreatic islet cell adaptation in obesity-associated tumors. Specifically, we identify aberrant beta cell expression of the peptide hormone cholecystokinin (Cck) in response to obesity and show that islet Cck promotes oncogenic Kras-driven pancreatic ductal tumorigenesis. Our studies argue that PDAC progression is driven by local obesity-associated changes in the tumor microenvironment and implicate endocrine-exocrine signaling beyond insulin in PDAC development.


Asunto(s)
Carcinoma Ductal Pancreático/etiología , Carcinoma Ductal Pancreático/metabolismo , Obesidad/metabolismo , Animales , Carcinogénesis/genética , Carcinoma Ductal Pancreático/patología , Línea Celular , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endocrinas/metabolismo , Glándulas Exocrinas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Obesidad/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Transducción de Señal/genética , Microambiente Tumoral/fisiología , Neoplasias Pancreáticas
8.
Cell ; 182(3): 563-577.e20, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32615086

RESUMEN

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.


Asunto(s)
Adipogénesis/genética , Tejido Adiposo Beige/metabolismo , Metabolismo Energético/genética , Quinasa 1 de Adhesión Focal/metabolismo , Transducción de Señal/genética , Células Madre/metabolismo , Tetraspanina 28/metabolismo , Adipocitos/metabolismo , Tejido Adiposo Beige/citología , Tejido Adiposo Beige/crecimiento & desarrollo , Tejido Adiposo Blanco/metabolismo , Adulto , Animales , Ataxina-1/metabolismo , Femenino , Fibronectinas/farmacología , Quinasa 1 de Adhesión Focal/genética , Humanos , Inflamación/genética , Inflamación/metabolismo , Resistencia a la Insulina/genética , Integrinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Obesidad/genética , Obesidad/metabolismo , RNA-Seq , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Análisis de la Célula Individual , Células Madre/citología , Tetraspanina 28/genética
9.
Cell ; 182(2): 270-296, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32707093

RESUMEN

Mammals have two specialized vascular circulatory systems: the blood vasculature and the lymphatic vasculature. The lymphatic vasculature is a unidirectional conduit that returns filtered interstitial arterial fluid and tissue metabolites to the blood circulation. It also plays major roles in immune cell trafficking and lipid absorption. As we discuss in this review, the molecular characterization of lymphatic vascular development and our understanding of this vasculature's role in pathophysiological conditions has greatly improved in recent years, changing conventional views about the roles of the lymphatic vasculature in health and disease. Morphological or functional defects in the lymphatic vasculature have now been uncovered in several pathological conditions. We propose that subtle asymptomatic alterations in lymphatic vascular function could underlie the variability seen in the body's response to a wide range of human diseases.


Asunto(s)
Vasos Linfáticos/metabolismo , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Historia del Siglo XXI , Humanos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Linfangiogénesis , Enfermedades Linfáticas/genética , Enfermedades Linfáticas/historia , Enfermedades Linfáticas/patología , Metástasis Linfática , Vasos Linfáticos/anatomía & histología , Vasos Linfáticos/citología , Neoplasias/metabolismo , Neoplasias/patología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética
10.
Cell ; 176(4): 729-742.e18, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30661757

RESUMEN

Hypothalamic melanocortin neurons play a pivotal role in weight regulation. Here, we examined the contribution of Semaphorin 3 (SEMA3) signaling to the development of these circuits. In genetic studies, we found 40 rare variants in SEMA3A-G and their receptors (PLXNA1-4; NRP1-2) in 573 severely obese individuals; variants disrupted secretion and/or signaling through multiple molecular mechanisms. Rare variants in this set of genes were significantly enriched in 982 severely obese cases compared to 4,449 controls. In a zebrafish mutagenesis screen, deletion of 7 genes in this pathway led to increased somatic growth and/or adiposity demonstrating that disruption of Semaphorin 3 signaling perturbs energy homeostasis. In mice, deletion of the Neuropilin-2 receptor in Pro-opiomelanocortin neurons disrupted their projections from the arcuate to the paraventricular nucleus, reduced energy expenditure, and caused weight gain. Cumulatively, these studies demonstrate that SEMA3-mediated signaling drives the development of hypothalamic melanocortin circuits involved in energy homeostasis.


Asunto(s)
Metabolismo Energético/genética , Melanocortinas/metabolismo , Semaforinas/genética , Adolescente , Adulto , Animales , Peso Corporal , Línea Celular , Niño , Preescolar , Modelos Animales de Enfermedad , Ingestión de Alimentos , Femenino , Variación Genética/genética , Homeostasis , Humanos , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Obesidad/genética , Obesidad/metabolismo , Receptores de Superficie Celular/metabolismo , Semaforinas/metabolismo , Adulto Joven , Pez Cebra
11.
Cell ; 179(6): 1289-1305.e21, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31761534

RESUMEN

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.


Asunto(s)
Adipogénesis , Cilios/metabolismo , Ácidos Grasos Omega-3/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Factor de Unión a CCCTC/metabolismo , Cromatina/metabolismo , Cilios/efectos de los fármacos , AMP Cíclico/metabolismo , Ácidos Docosahexaenoicos/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , PPAR gamma/metabolismo
12.
Cell ; 177(3): 597-607.e9, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-31002796

RESUMEN

The melanocortin 4 receptor (MC4R) is a G protein-coupled receptor whose disruption causes obesity. We functionally characterized 61 MC4R variants identified in 0.5 million people from UK Biobank and examined their associations with body mass index (BMI) and obesity-related cardiometabolic diseases. We found that the maximal efficacy of ß-arrestin recruitment to MC4R, rather than canonical Gαs-mediated cyclic adenosine-monophosphate production, explained 88% of the variance in the association of MC4R variants with BMI. While most MC4R variants caused loss of function, a subset caused gain of function; these variants were associated with significantly lower BMI and lower odds of obesity, type 2 diabetes, and coronary artery disease. Protective associations were driven by MC4R variants exhibiting signaling bias toward ß-arrestin recruitment and increased mitogen-activated protein kinase pathway activation. Harnessing ß-arrestin-biased MC4R signaling may represent an effective strategy for weight loss and the treatment of obesity-related cardiometabolic diseases.


Asunto(s)
Mutación con Ganancia de Función/genética , Obesidad/patología , Receptor de Melanocortina Tipo 4/genética , Transducción de Señal , Adulto , Anciano , Índice de Masa Corporal , Enfermedad de la Arteria Coronaria/complicaciones , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , AMP Cíclico/metabolismo , Bases de Datos Factuales , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Obesidad/complicaciones , Obesidad/metabolismo , Polimorfismo de Nucleótido Simple , Receptor de Melanocortina Tipo 4/química , Receptor de Melanocortina Tipo 4/metabolismo , beta-Arrestinas/metabolismo
13.
Cell ; 178(3): 686-698.e14, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31257031

RESUMEN

Immune cells residing in white adipose tissue have been highlighted as important factors contributing to the pathogenesis of metabolic diseases, but the molecular regulators that drive adipose tissue immune cell remodeling during obesity remain largely unknown. Using index and transcriptional single-cell sorting, we comprehensively map all adipose tissue immune populations in both mice and humans during obesity. We describe a novel and conserved Trem2+ lipid-associated macrophage (LAM) subset and identify markers, spatial localization, origin, and functional pathways associated with these cells. Genetic ablation of Trem2 in mice globally inhibits the downstream molecular LAM program, leading to adipocyte hypertrophy as well as systemic hypercholesterolemia, body fat accumulation, and glucose intolerance. These findings identify Trem2 signaling as a major pathway by which macrophages respond to loss of tissue-level lipid homeostasis, highlighting Trem2 as a key sensor of metabolic pathologies across multiple tissues and a potential therapeutic target in metabolic diseases.


Asunto(s)
Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/metabolismo , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Animales , Dieta Alta en Grasa , Intolerancia a la Glucosa , Humanos , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Metabolismo de los Lípidos/genética , Lípidos/análisis , Macrófagos/citología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Monocitos/metabolismo , Obesidad/metabolismo , Obesidad/patología , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Transducción de Señal , Análisis de la Célula Individual
14.
Cell ; 177(3): 587-596.e9, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-31002795

RESUMEN

Severe obesity is a rapidly growing global health threat. Although often attributed to unhealthy lifestyle choices or environmental factors, obesity is known to be heritable and highly polygenic; the majority of inherited susceptibility is related to the cumulative effect of many common DNA variants. Here we derive and validate a new polygenic predictor comprised of 2.1 million common variants to quantify this susceptibility and test this predictor in more than 300,000 individuals ranging from middle age to birth. Among middle-aged adults, we observe a 13-kg gradient in weight and a 25-fold gradient in risk of severe obesity across polygenic score deciles. In a longitudinal birth cohort, we note minimal differences in birthweight across score deciles, but a significant gradient emerged in early childhood and reached 12 kg by 18 years of age. This new approach to quantify inherited susceptibility to obesity affords new opportunities for clinical prevention and mechanistic assessment.


Asunto(s)
Peso Corporal , Herencia Multifactorial/genética , Obesidad/patología , Adolescente , Índice de Masa Corporal , Niño , Bases de Datos Factuales , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Recién Nacido , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Obesidad/genética , Factores de Riesgo , Índice de Severidad de la Enfermedad
15.
Annu Rev Cell Dev Biol ; 36: 551-574, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-33021823

RESUMEN

Innate and adaptive immune responses decline with age, leading to greater susceptibility to infectious diseases and reduced responses to vaccines. Diseases are more severe in old than in young individuals and have a greater impact on health outcomes such as morbidity, disability, and mortality. Aging is characterized by increased low-grade chronic inflammation, so-called inflammaging, that represents a link between changes in immune cells and a number of diseases and syndromes typical of old age. In this review we summarize current knowledge on age-associated changes in immune cells with special emphasis on B cells, which are more inflammatory and less responsive to infections and vaccines in the elderly. We highlight recent findings on factors and pathways contributing to inflammaging and how these lead to dysfunctional immune responses. We summarize recent published studies showing that adipose tissue, which increases in size with aging, contributes to inflammaging and dysregulated B cell function.


Asunto(s)
Linfocitos B/citología , Linfocitos B/inmunología , Inmunosenescencia , Animales , Formación de Anticuerpos/inmunología , Microbioma Gastrointestinal/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Polimorfismo de Nucleótido Simple/genética
16.
Immunity ; 57(1): 141-152.e5, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38091996

RESUMEN

Adipose tissues (ATs) are innervated by sympathetic nerves, which drive reduction of fat mass via lipolysis and thermogenesis. Here, we report a population of immunomodulatory leptin receptor-positive (LepR+) sympathetic perineurial barrier cells (SPCs) present in mice and humans, which uniquely co-express Lepr and interleukin-33 (Il33) and ensheath AT sympathetic axon bundles. Brown ATs (BATs) of mice lacking IL-33 in SPCs (SPCΔIl33) had fewer regulatory T (Treg) cells and eosinophils, resulting in increased BAT inflammation. SPCΔIl33 mice were more susceptible to diet-induced obesity, independently of food intake. Furthermore, SPCΔIl33 mice had impaired adaptive thermogenesis and were unresponsive to leptin-induced rescue of metabolic adaptation. We therefore identify LepR+ SPCs as a source of IL-33, which orchestrate an anti-inflammatory BAT environment, preserving sympathetic-mediated thermogenesis and body weight homeostasis. LepR+IL-33+ SPCs provide a cellular link between leptin and immune regulation of body weight, unifying neuroendocrinology and immunometabolism as previously disconnected fields of obesity research.


Asunto(s)
Tejido Adiposo Pardo , Leptina , Animales , Humanos , Ratones , Tejido Adiposo Pardo/inervación , Tejido Adiposo Pardo/metabolismo , Peso Corporal , Metabolismo Energético/fisiología , Interleucina-33/genética , Interleucina-33/metabolismo , Obesidad/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Termogénesis/fisiología
17.
Cell ; 175(3): 605-614, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30340032

RESUMEN

Modern nutrition is often characterized by the excessive intake of different types of carbohydrates ranging from digestible polysaccharides to refined sugars that collectively mediate noxious effects on human health, a phenomenon that we refer to as "carbotoxicity." Epidemiological and experimental evidence combined with clinical intervention trials underscore the negative impact of excessive carbohydrate uptake, as well as the beneficial effects of reducing carbs in the diet. We discuss the molecular, cellular, and neuroendocrine mechanisms that link exaggerated carbohydrate intake to disease and accelerated aging as we outline dietary and pharmacologic strategies to combat carbotoxicity.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Carbohidratos de la Dieta/efectos adversos , Animales , Metabolismo de los Hidratos de Carbono , Cardiotoxicidad , Humanos
18.
Cell ; 175(4): 962-972.e10, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388453

RESUMEN

Many US immigrant populations develop metabolic diseases post immigration, but the causes are not well understood. Although the microbiome plays a role in metabolic disease, there have been no studies measuring the effects of US immigration on the gut microbiome. We collected stool, dietary recalls, and anthropometrics from 514 Hmong and Karen individuals living in Thailand and the United States, including first- and second-generation immigrants and 19 Karen individuals sampled before and after immigration, as well as from 36 US-born European American individuals. Using 16S and deep shotgun metagenomic DNA sequencing, we found that migration from a non-Western country to the United States is associated with immediate loss of gut microbiome diversity and function in which US-associated strains and functions displace native strains and functions. These effects increase with duration of US residence and are compounded by obesity and across generations.


Asunto(s)
Pueblo Asiatico , Emigración e Inmigración , Microbioma Gastrointestinal , Adulto , Bacteroides/aislamiento & purificación , Fibras de la Dieta/metabolismo , Emigrantes e Inmigrantes , Humanos , Metagenoma , Obesidad/epidemiología , Obesidad/microbiología , Prevotella/aislamiento & purificación , Estados Unidos
19.
Cell ; 175(3): 695-708.e13, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30293865

RESUMEN

We have uncovered the existence of extracellular vesicle (EV)-mediated signaling between cell types within the adipose tissue (AT) proper. This phenomenon became evident in our attempts at generating an adipocyte-specific knockout of caveolin 1 (cav1) protein. Although we effectively ablated the CAV1 gene in adipocytes, cav1 protein remained abundant. With the use of newly generated mouse models, we show that neighboring endothelial cells (ECs) transfer cav1-containing EVs to adipocytes in vivo, which reciprocate by releasing EVs to ECs. AT-derived EVs contain proteins and lipids capable of modulating cellular signaling pathways. Furthermore, this mechanism facilitates transfer of plasma constituents from ECs to the adipocyte. The transfer event is physiologically regulated by fasting/refeeding and obesity, suggesting EVs participate in the tissue response to changes in the systemic nutrient state. This work offers new insights into the complex signaling mechanisms that exist among adipocytes, stromal vascular cells, and, potentially, distal organs.


Asunto(s)
Adipocitos/metabolismo , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Ayuno/metabolismo , Transducción de Señal , Animales , Caveolina 1/genética , Caveolina 1/metabolismo , Línea Celular , Células Cultivadas , Endotelio Vascular/citología , Masculino , Ratones , Ratones Endogámicos C57BL
20.
Cell ; 174(4): 831-842.e12, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30057115

RESUMEN

Overnutrition disrupts circadian metabolic rhythms by mechanisms that are not well understood. Here, we show that diet-induced obesity (DIO) causes massive remodeling of circadian enhancer activity in mouse liver, triggering synchronous high-amplitude circadian rhythms of both fatty acid (FA) synthesis and oxidation. SREBP expression was rhythmically induced by DIO, leading to circadian FA synthesis and, surprisingly, FA oxidation (FAO). DIO similarly caused a high-amplitude circadian rhythm of PPARα, which was also required for FAO. Provision of a pharmacological activator of PPARα abrogated the requirement of SREBP for FAO (but not FA synthesis), suggesting that SREBP indirectly controls FAO via production of endogenous PPARα ligands. The high-amplitude rhythm of PPARα imparted time-of-day-dependent responsiveness to lipid-lowering drugs. Thus, acquisition of rhythmicity for non-core clock components PPARα and SREBP1 remodels metabolic gene transcription in response to overnutrition and enables a chronopharmacological approach to metabolic disorders.


Asunto(s)
Ritmo Circadiano , Dieta/efectos adversos , Hígado/metabolismo , Obesidad/metabolismo , PPAR alfa/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Animales , Regulación de la Expresión Génica , Metabolismo de los Lípidos , Lipogénesis , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/patología , PPAR alfa/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA