Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(10): 2359-2374.e18, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38653240

RESUMEN

Brown adipose tissue (BAT) is best known for thermogenesis. Rodent studies demonstrated that enhanced BAT thermogenesis is tightly associated with increased energy expenditure, reduced body weight, and improved glucose homeostasis. However, human BAT is protective against type 2 diabetes, independent of body weight. The mechanism underlying this dissociation remains unclear. Here, we report that impaired mitochondrial catabolism of branched-chain amino acids (BCAAs) in BAT, by deleting mitochondrial BCAA carriers (MBCs), caused systemic insulin resistance without affecting energy expenditure and body weight. Brown adipocytes catabolized BCAA in the mitochondria as nitrogen donors for the biosynthesis of non-essential amino acids and glutathione. Impaired mitochondrial BCAA-nitrogen flux in BAT resulted in increased oxidative stress, decreased hepatic insulin signaling, and decreased circulating BCAA-derived metabolites. A high-fat diet attenuated BCAA-nitrogen flux and metabolite synthesis in BAT, whereas cold-activated BAT enhanced the synthesis. This work uncovers a metabolite-mediated pathway through which BAT controls metabolic health beyond thermogenesis.


Asunto(s)
Tejido Adiposo Pardo , Aminoácidos de Cadena Ramificada , Resistencia a la Insulina , Mitocondrias , Nitrógeno , Termogénesis , Tejido Adiposo Pardo/metabolismo , Animales , Aminoácidos de Cadena Ramificada/metabolismo , Ratones , Nitrógeno/metabolismo , Mitocondrias/metabolismo , Masculino , Humanos , Metabolismo Energético , Ratones Endogámicos C57BL , Estrés Oxidativo , Insulina/metabolismo , Dieta Alta en Grasa , Adipocitos Marrones/metabolismo , Transducción de Señal
2.
Cell ; 185(24): 4654-4673.e28, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-36334589

RESUMEN

Brown adipose tissue (BAT) regulates metabolic physiology. However, nearly all mechanistic studies of BAT protein function occur in a single inbred mouse strain, which has limited the understanding of generalizable mechanisms of BAT regulation over physiology. Here, we perform deep quantitative proteomics of BAT across a cohort of 163 genetically defined diversity outbred mice, a model that parallels the genetic and phenotypic variation found in humans. We leverage this diversity to define the functional architecture of the outbred BAT proteome, comprising 10,479 proteins. We assign co-operative functions to 2,578 proteins, enabling systematic discovery of regulators of BAT. We also identify 638 proteins that correlate with protection from, or sensitivity to, at least one parameter of metabolic disease. We use these findings to uncover SFXN5, LETMD1, and ATP1A2 as modulators of BAT thermogenesis or adiposity, and provide OPABAT as a resource for understanding the conserved mechanisms of BAT regulation over metabolic physiology.


Asunto(s)
Tejido Adiposo Pardo , Proteoma , Humanos , Ratones , Animales , Tejido Adiposo Pardo/metabolismo , Proteoma/metabolismo , Termogénesis/fisiología , Adiposidad , Obesidad/metabolismo , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas/metabolismo
3.
Nat Immunol ; 20(3): 373, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30728493

RESUMEN

In the version of this article initially published, three authors (Hui-Fern Kuoy, Adam P. Uldrich and Dale. I. Godfrey) and their affiliations, acknowledgments and contributions were not included. The correct information is as follows:Ayano C. Kohlgruber1,2, Shani T. Gal-Oz3, Nelson M. LaMarche1,2, Moto Shimazaki1, Danielle Duquette4, Hui-Fern Koay5,6, Hung N. Nguyen1, Amir I. Mina4, Tyler Paras1, Ali Tavakkoli7, Ulrich von Andrian2,8, Adam P. Uldrich5,6, Dale I. Godfrey5,6, Alexander S. Banks4, Tal Shay3, Michael B. Brenner1,10* and Lydia Lynch1,4,9,10*1Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA, USA. 2Division of Medical Sciences, Harvard Medical School, Boston, MA, USA. 3Department of Life Sciences, Ben-Gurion University of the Negev, Beersheba, Israel. 4Division of Endocrinology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA. 5Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Australia. 6ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Australia. 7Department of General and Gastrointestinal Surgery, Brigham and Women's Hospital, Boston, MA, USA. 8Department of Microbiology and Immunology, Harvard Medical School, Boston, MA, USA. 9School of Biochemistry and Immunology, Trinity College, Dublin, Ireland. 10These authors jointly supervised this work: Michael B. Brenner, Lydia Lynch. *e-mail: mbrenner@research.bwh.harvard.edu; llynch@bwh.harvard.eduAcknowledgementsWe thank A.T. Chicoine, flow cytometry core manager at the Human Immunology Center at BWH, for flow cytometry sorting. We thank D. Sant'Angelo (Rutgers Cancer Institute) for providing Zbtb16-/- mice and R. O'Brien (National Jewish Health) for providing Vg4/6-/- mice. Supported by NIH grant R01 AI11304603 (to M.B.B.), ERC Starting Grant 679173 (to L.L.), the National Health and Medical Research Council of Australia (1013667), an Australian Research Council Future Fellowship (FT140100278 for A.P.U.) and a National Health and Medical Research Council of Australia Senior Principal Research Fellowship (1117766 for D.I.G.).Author contributionsA.C.K., L.L., and M.B.B. conceived and designed the experiments, and wrote the manuscript. A.C.K., N.M.L., L.L., H.N.N., M.S., T.P., and D.D. performed the experiments. S.T.G.-O. and T.S. performed the RNA-seq analysis. A.S.B. and A.I.M. provided advice and performed the CLAMS experiments. A.T. provided human bariatric patient samples. Parabiosis experiments were performed in the laboratory of U.v.A. H.-F.K., A.P.U. and D.I.G provided critical insight into the TCR chain usage of PLZF+ γδ T cells. M.B.B., N.M.L., and L.L. critically reviewed the manuscript.The errors have been corrected in the HTML and PDF version of the article.Correction to: Nature Immunology doi:10.1038/s41590-018-0094-2 (2018), published online 18 April 2018.

4.
Nat Immunol ; 19(5): 464-474, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29670241

RESUMEN

γδ T cells are situated at barrier sites and guard the body from infection and damage. However, little is known about their roles outside of host defense in nonbarrier tissues. Here, we characterize a highly enriched tissue-resident population of γδ T cells in adipose tissue that regulate age-dependent regulatory T cell (Treg) expansion and control core body temperature in response to environmental fluctuations. Mechanistically, innate PLZF+ γδ T cells produced tumor necrosis factor and interleukin (IL) 17 A and determined PDGFRα+ and Pdpn+ stromal-cell production of IL-33 in adipose tissue. Mice lacking γδ T cells or IL-17A exhibited decreases in both ST2+ Treg cells and IL-33 abundance in visceral adipose tissue. Remarkably, these mice also lacked the ability to regulate core body temperature at thermoneutrality and after cold challenge. Together, these findings uncover important physiological roles for resident γδ T cells in adipose tissue immune homeostasis and body-temperature control.


Asunto(s)
Tejido Adiposo/citología , Homeostasis/fisiología , Interleucina-17/metabolismo , Linfocitos T Reguladores/fisiología , Termogénesis/fisiología , Tejido Adiposo/fisiología , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos T gamma-delta , Subgrupos de Linfocitos T/fisiología
5.
Cell ; 158(1): 41-53, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24995977

RESUMEN

A hallmark of type 2 diabetes mellitus (T2DM) is the development of pancreatic ß cell failure, which results in insulinopenia and hyperglycemia. We show that the adipokine adipsin has a beneficial role in maintaining ß cell function. Animals genetically lacking adipsin have glucose intolerance due to insulinopenia; isolated islets from these mice have reduced glucose-stimulated insulin secretion. Replenishment of adipsin to diabetic mice treated hyperglycemia by boosting insulin secretion. We identify C3a, a peptide generated by adipsin, as a potent insulin secretagogue and show that the C3a receptor is required for these beneficial effects of adipsin. C3a acts on islets by augmenting ATP levels, respiration, and cytosolic free Ca(2+). Finally, we demonstrate that T2DM patients with ß cell failure are deficient in adipsin. These findings indicate that the adipsin/C3a pathway connects adipocyte function to ß cell physiology, and manipulation of this molecular switch may serve as a therapy in T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Tejido Adiposo/metabolismo , Animales , Complemento C3a/metabolismo , Factor D del Complemento/genética , Factor D del Complemento/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Dieta Alta en Grasa , Glucosa/metabolismo , Humanos , Inflamación/metabolismo , Insulina/metabolismo , Secreción de Insulina , Ratones
6.
Cell ; 156(1-2): 304-16, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24439384

RESUMEN

A clear relationship exists between visceral obesity and type 2 diabetes, whereas subcutaneous obesity is comparatively benign. Here, we show that adipocyte-specific deletion of the coregulatory protein PRDM16 caused minimal effects on classical brown fat but markedly inhibited beige adipocyte function in subcutaneous fat following cold exposure or ß3-agonist treatment. These animals developed obesity on a high-fat diet, with severe insulin resistance and hepatic steatosis. They also showed altered fat distribution with markedly increased subcutaneous adiposity. Subcutaneous adipose tissue in mutant mice acquired many key properties of visceral fat, including decreased thermogenic and increased inflammatory gene expression and increased macrophage accumulation. Transplantation of subcutaneous fat into mice with diet-induced obesity showed a loss of metabolic benefit when tissues were derived from PRDM16 mutant animals. These findings indicate that PRDM16 and beige adipocytes are required for the "browning" of white fat and the healthful effects of subcutaneous adipose tissue.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo/metabolismo , Proteínas de Unión al ADN/metabolismo , Obesidad/metabolismo , Factores de Transcripción/metabolismo , Adipocitos/metabolismo , Animales , Proteínas de Unión al ADN/genética , Dieta Alta en Grasa , Resistencia a la Insulina , Ratones , Ratones Noqueados , Factores de Transcripción/genética
7.
Nature ; 621(7977): 138-145, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37587337

RESUMEN

Maintaining body temperature is calorically expensive for endothermic animals1. Mammals eat more in the cold to compensate for energy expenditure2, but the neural mechanism underlying this coupling is not well understood. Through behavioural and metabolic analyses, we found that mice dynamically switch between energy-conservation and food-seeking states in the cold, the latter of which are primarily driven by energy expenditure rather than the sensation of cold. To identify the neural mechanisms underlying cold-induced food seeking, we used whole-brain c-Fos mapping and found that the xiphoid (Xi), a small nucleus in the midline thalamus, was selectively activated by prolonged cold associated with elevated energy expenditure but not with acute cold exposure. In vivo calcium imaging showed that Xi activity correlates with food-seeking episodes under cold conditions. Using activity-dependent viral strategies, we found that optogenetic and chemogenetic stimulation of cold-activated Xi neurons selectively recapitulated food seeking under cold conditions whereas their inhibition suppressed it. Mechanistically, Xi encodes a context-dependent valence switch that promotes food-seeking behaviours under cold but not warm conditions. Furthermore, these behaviours are mediated by a Xi-to-nucleus accumbens projection. Our results establish Xi as a key region in the control of cold-induced feeding, which is an important mechanism in the maintenance of energy homeostasis in endothermic animals.


Asunto(s)
Temperatura Corporal , Frío , Conducta Alimentaria , Tálamo , Animales , Ratones , Temperatura Corporal/fisiología , Mapeo Encefálico , Calcio/metabolismo , Conducta Alimentaria/fisiología , Metabolismo Energético/fisiología , Tálamo/anatomía & histología , Tálamo/citología , Tálamo/fisiología , Optogenética , Neuronas/metabolismo , Núcleo Accumbens/citología , Núcleo Accumbens/fisiología , Homeostasis/fisiología , Termogénesis/fisiología
8.
Cell ; 151(1): 96-110, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-23021218

RESUMEN

PGC1α is a key transcriptional coregulator of oxidative metabolism and thermogenesis. Through a high-throughput chemical screen, we found that molecules antagonizing the TRPVs (transient receptor potential vanilloid), a family of ion channels, induced PGC1α expression in adipocytes. In particular, TRPV4 negatively regulated the expression of PGC1α, UCP1, and cellular respiration. Additionally, it potently controlled the expression of multiple proinflammatory genes involved in the development of insulin resistance. Mice with a null mutation for TRPV4 or wild-type mice treated with a TRPV4 antagonist showed elevated thermogenesis in adipose tissues and were protected from diet-induced obesity, adipose inflammation, and insulin resistance. This role of TRPV4 as a cell-autonomous mediator for both the thermogenic and proinflammatory programs in adipocytes could offer a target for treating obesity and related metabolic diseases.


Asunto(s)
Metabolismo Energético , Canales Catiónicos TRPV/metabolismo , Termogénesis , Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Femenino , Técnicas de Silenciamiento del Gen , Canales Iónicos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/metabolismo , Obesidad/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética , Transactivadores/metabolismo , Factores de Transcripción , Proteína Desacopladora 1
9.
Nature ; 578(7796): 610-614, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32076265

RESUMEN

The sympathetic nervous system innervates peripheral organs to regulate their function and maintain homeostasis, whereas target cells also produce neurotrophic factors to promote sympathetic innervation1,2. The molecular basis of this bi-directional communication remains to be fully determined. Here we use thermogenic adipose tissue from mice as a model system to show that T cells, specifically γδ T cells, have a crucial role in promoting sympathetic innervation, at least in part by driving the expression of TGFß1 in parenchymal cells via the IL-17 receptor C (IL-17RC). Ablation of IL-17RC specifically in adipose tissue reduces expression of TGFß1 in adipocytes, impairs local sympathetic innervation and causes obesity and other metabolic phenotypes that are consistent with defective thermogenesis; innervation can be fully rescued by restoring TGFß1 expression. Ablating γδ Τ cells and the IL-17RC signalling pathway also impairs sympathetic innervation in other tissues such as salivary glands. These findings demonstrate coordination between T cells and parenchymal cells to regulate sympathetic innervation.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/inervación , Tejido Adiposo/metabolismo , Interleucina-17/metabolismo , Sistema Nervioso Simpático/fisiología , Linfocitos T/metabolismo , Termogénesis , Tejido Adiposo Pardo/metabolismo , Animales , Interleucina-17/deficiencia , Interleucina-17/genética , Masculino , Ratones , Ratones Noqueados , Especificidad de Órganos , Tejido Parenquimatoso/citología , Transducción de Señal , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
10.
Nature ; 583(7814): 115-121, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32528180

RESUMEN

The advent of endothermy, which is achieved through the continuous homeostatic regulation of body temperature and metabolism1,2, is a defining feature of mammalian and avian evolution. However, when challenged by food deprivation or harsh environmental conditions, many mammalian species initiate adaptive energy-conserving survival strategies-including torpor and hibernation-during which their body temperature decreases far below its homeostatic set-point3-5. How homeothermic mammals initiate and regulate these hypothermic states remains largely unknown. Here we show that entry into mouse torpor, a fasting-induced state with a greatly decreased metabolic rate and a body temperature as low as 20 °C6, is regulated by neurons in the medial and lateral preoptic area of the hypothalamus. We show that restimulation of neurons that were activated during a previous bout of torpor is sufficient to initiate the key features of torpor, even in mice that are not calorically restricted. Among these neurons we identify a population of glutamatergic Adcyap1-positive cells, the activity of which accurately determines when mice naturally initiate and exit torpor, and the inhibition of which disrupts the natural process of torpor entry, maintenance and arousal. Taken together, our results reveal a specific neuronal population in the mouse hypothalamus that serves as a core regulator of torpor. This work forms a basis for the future exploration of mechanisms and circuitry that regulate extreme hypothermic and hypometabolic states, and enables genetic access to monitor, initiate, manipulate and study these ancient adaptations of homeotherm biology.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/citología , Vías Nerviosas/fisiología , Neuronas/fisiología , Letargo/fisiología , Animales , Ayuno , Femenino , Privación de Alimentos , Glutamina/metabolismo , Hipotálamo/fisiología , Masculino , Ratones , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo
11.
Nature ; 560(7716): 102-106, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30022159

RESUMEN

Thermogenesis by brown and beige adipose tissue, which requires activation by external stimuli, can counter metabolic disease1. Thermogenic respiration is initiated by adipocyte lipolysis through cyclic AMP-protein kinase A signalling; this pathway has been subject to longstanding clinical investigation2-4. Here we apply a comparative metabolomics approach and identify an independent metabolic pathway that controls acute activation of adipose tissue thermogenesis in vivo. We show that substantial and selective accumulation of the tricarboxylic acid cycle intermediate succinate is a metabolic signature of adipose tissue thermogenesis upon activation by exposure to cold. Succinate accumulation occurs independently of adrenergic signalling, and is sufficient to elevate thermogenic respiration in brown adipocytes. Selective accumulation of succinate may be driven by a capacity of brown adipocytes to sequester elevated circulating succinate. Furthermore, brown adipose tissue thermogenesis can be initiated by systemic administration of succinate in mice. Succinate from the extracellular milieu is rapidly metabolized by brown adipocytes, and its oxidation by succinate dehydrogenase is required for activation of thermogenesis. We identify a mechanism whereby succinate dehydrogenase-mediated oxidation of succinate initiates production of reactive oxygen species, and drives thermogenic respiration, whereas inhibition of succinate dehydrogenase supresses thermogenesis. Finally, we show that pharmacological elevation of circulating succinate drives UCP1-dependent thermogenesis by brown adipose tissue in vivo, which stimulates robust protection against diet-induced obesity and improves glucose tolerance. These findings reveal an unexpected mechanism for control of thermogenesis, using succinate as a systemically-derived thermogenic molecule.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Ácido Succínico/metabolismo , Termogénesis/fisiología , Adipocitos/efectos de los fármacos , Adipocitos/enzimología , Adipocitos/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/enzimología , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/enzimología , Tejido Adiposo Blanco/metabolismo , Animales , Femenino , Masculino , Metabolómica , Ratones , Obesidad/metabolismo , Obesidad/prevención & control , Oxidación-Reducción/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Succinato Deshidrogenasa/metabolismo , Ácido Succínico/farmacología , Termogénesis/efectos de los fármacos , Proteína Desacopladora 1/metabolismo
12.
Nat Chem Biol ; 16(3): 318-326, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32042200

RESUMEN

Bile salt hydrolase (BSH) enzymes are widely expressed by human gut bacteria and catalyze the gateway reaction leading to secondary bile acid formation. Bile acids regulate key metabolic and immune processes by binding to host receptors. There is an unmet need for a potent tool to inhibit BSHs across all gut bacteria to study the effects of bile acids on host physiology. Here, we report the development of a covalent pan-inhibitor of gut bacterial BSHs. From a rationally designed candidate library, we identified a lead compound bearing an alpha-fluoromethyl ketone warhead that modifies BSH at the catalytic cysteine residue. This inhibitor abolished BSH activity in conventional mouse feces. Mice gavaged with a single dose of this compound displayed decreased BSH activity and decreased deconjugated bile acid levels in feces. Our studies demonstrate the potential of a covalent BSH inhibitor to modulate bile acid composition in vivo.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Amidohidrolasas/metabolismo , Microbioma Gastrointestinal/fisiología , Amidohidrolasas/fisiología , Animales , Bacterias/enzimología , Ácidos y Sales Biliares/metabolismo , Diseño de Fármacos , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Bibliotecas de Moléculas Pequeñas
13.
Genes Dev ; 28(21): 2361-9, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25316675

RESUMEN

Phosphorylation of peroxisome proliferator-activated receptor γ (PPARγ) at Ser273 by cyclin-dependent kinase 5 (CDK5) in adipose tissue stimulates insulin resistance, but the underlying molecular mechanisms are unclear. We show here that Thrap3 (thyroid hormone receptor-associated protein 3) can directly interact with PPARγ when it is phosphorylated at Ser273, and this interaction controls the diabetic gene programming mediated by the phosphorylation of PPARγ. Knockdown of Thrap3 restores most of the genes dysregulated by CDK5 action on PPARγ in cultured adipocytes. Importantly, reduced expression of Thrap3 in fat tissue by antisense oligonucleotides (ASOs) regulates a specific set of genes, including the key adipokines adiponectin and adipsin, and effectively improves hyperglycemia and insulin resistance in high-fat-fed mice without affecting body weight. These data indicate that Thrap3 plays a crucial role in controlling diabetic gene programming and may provide opportunities for the development of new therapeutics for obesity and type 2 diabetes.


Asunto(s)
Proteínas de Unión al ADN/genética , Diabetes Mellitus Tipo 2/genética , PPAR gamma/metabolismo , Factores de Transcripción/genética , Células 3T3 , Adipoquinas/genética , Animales , Células Cultivadas , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Fosfoserina/metabolismo , Unión Proteica
14.
Nature ; 517(7534): 391-5, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25409143

RESUMEN

Obesity-linked insulin resistance is a major precursor to the development of type 2 diabetes. Previous work has shown that phosphorylation of PPARγ (peroxisome proliferator-activated receptor γ) at serine 273 by cyclin-dependent kinase 5 (Cdk5) stimulates diabetogenic gene expression in adipose tissues. Inhibition of this modification is a key therapeutic mechanism for anti-diabetic drugs that bind PPARγ, such as the thiazolidinediones and PPARγ partial agonists or non-agonists. For a better understanding of the importance of this obesity-linked PPARγ phosphorylation, we created mice that ablated Cdk5 specifically in adipose tissues. These mice have both a paradoxical increase in PPARγ phosphorylation at serine 273 and worsened insulin resistance. Unbiased proteomic studies show that extracellular signal-regulated kinase (ERK) kinases are activated in these knockout animals. Here we show that ERK directly phosphorylates serine 273 of PPARγ in a robust manner and that Cdk5 suppresses ERKs through direct action on a novel site in MAP kinase/ERK kinase (MEK). Importantly, pharmacological inhibition of MEK and ERK markedly improves insulin resistance in both obese wild-type and ob/ob mice, and also completely reverses the deleterious effects of the Cdk5 ablation. These data show that an ERK/Cdk5 axis controls PPARγ function and suggest that MEK/ERK inhibitors may hold promise for the treatment of type 2 diabetes.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Diabetes Mellitus/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , PPAR gamma/metabolismo , Adipocitos/enzimología , Adipocitos/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/enzimología , Tejido Adiposo/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Quinasa 5 Dependiente de la Ciclina/deficiencia , Dieta Alta en Grasa , Humanos , Resistencia a la Insulina , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , PPAR gamma/química , Fosforilación
15.
Proc Natl Acad Sci U S A ; 115(3): 561-566, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29295932

RESUMEN

The peroxisome-proliferator receptor-γ (PPARγ) is expressed in multiple cancer types. Recently, our group has shown that PPARγ is phosphorylated on serine 273 (S273), which selectively modulates the transcriptional program controlled by this protein. PPARγ ligands, including thiazolidinediones (TZDs), block S273 phosphorylation. This activity is chemically separable from the canonical activation of the receptor by agonist ligands and, importantly, these noncanonical agonist ligands do not cause some of the known side effects of TZDs. Here, we show that phosphorylation of S273 of PPARγ occurs in cancer cells on exposure to DNA damaging agents. Blocking this phosphorylation genetically or pharmacologically increases accumulation of DNA damage, resulting in apoptotic cell death. A genetic signature of PPARγ phosphorylation is associated with worse outcomes in response to chemotherapy in human patients. Noncanonical agonist ligands sensitize lung cancer xenografts and genetically induced lung tumors to carboplatin therapy. Moreover, inhibition of this phosphorylation results in deregulation of p53 signaling, and biochemical studies show that PPARγ physically interacts with p53 in a manner dependent on S273 phosphorylation. These data implicate a role for PPARγ in modifying the p53 response to cytotoxic therapy, which can be modulated for therapeutic gain using these compounds.


Asunto(s)
Antineoplásicos/administración & dosificación , Daño del ADN , Neoplasias Pulmonares/tratamiento farmacológico , PPAR gamma/metabolismo , Tiazolidinedionas/administración & dosificación , Secuencias de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Carboplatino/administración & dosificación , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Humanos , Ligandos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Desnudos , PPAR gamma/agonistas , PPAR gamma/química , PPAR gamma/genética , Fosforilación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
16.
Proc Natl Acad Sci U S A ; 115(29): E6937-E6945, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-29967167

RESUMEN

N-acyl amino acids (NAAs) are a structurally diverse class of bioactive signaling lipids whose endogenous functions have largely remained uncharacterized. To clarify the physiologic roles of NAAs, we generated mice deficient in the circulating enzyme peptidase M20 domain-containing 1 (PM20D1). Global PM20D1-KO mice have dramatically reduced NAA hydrolase/synthase activities in tissues and blood with concomitant bidirectional dysregulation of endogenous NAAs. Compared with control animals, PM20D1-KO mice exhibit a variety of metabolic and pain phenotypes, including insulin resistance, altered body temperature in cold, and antinociceptive behaviors. Guided by these phenotypes, we identify N-oleoyl-glutamine (C18:1-Gln) as a key PM20D1-regulated NAA. In addition to its mitochondrial uncoupling bioactivity, C18:1-Gln also antagonizes certain members of the transient receptor potential (TRP) calcium channels including TRPV1. Direct administration of C18:1-Gln to mice is sufficient to recapitulate a subset of phenotypes observed in PM20D1-KO animals. These data demonstrate that PM20D1 is a dominant enzymatic regulator of NAA levels in vivo and elucidate physiologic functions for NAA signaling in metabolism and nociception.


Asunto(s)
Amidohidrolasas/metabolismo , Glutamina/metabolismo , Nocicepción/fisiología , Ácidos Oléicos/metabolismo , Transducción de Señal/fisiología , Amidohidrolasas/genética , Animales , Temperatura Corporal/fisiología , Glutamina/genética , Glutamina/farmacología , Ratones , Ratones Noqueados , Nocicepción/efectos de los fármacos , Ácidos Oléicos/genética , Ácidos Oléicos/farmacología , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo
17.
Am J Physiol Endocrinol Metab ; 318(5): E678-E688, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32069072

RESUMEN

Sleeve gastrectomy (SG) induces weight loss-independent improvements in glucose homeostasis by unknown mechanisms. We sought to identify the metabolic adaptations responsible for these improvements. Nonobese C57BL/6J mice on standard chow underwent SG or sham surgery. Functional testing and indirect calorimetry were used to capture metabolic phenotypes. Tissue-specific glucose uptake was assessed by 18-fluorodeoxyglucose (18-FDG) PET/computed tomography, and RNA sequencing was used for gene-expression analysis. In this model, SG induced durable improvements in glucose tolerance in the absence of changes in weight, body composition, or food intake. Indirect calorimetry revealed that SG increased the average respiratory exchange ratio toward 1.0, indicating a weight-independent, systemic shift to carbohydrate utilization. Following SG, orally administered 18-FDG preferentially localized to white adipose depots, showing tissue-specific increases in glucose utilization induced by surgery. Transcriptional analysis with RNA sequencing demonstrated that increased glucose uptake in the visceral adipose tissue was associated with upregulation in transcriptional pathways involved in energy metabolism, adipocyte maturation, and adaptive and innate immune cell chemotaxis and differentiation. SG induces a rapid, weight loss-independent shift toward glucose utilization and transcriptional remodeling of metabolic and immune pathways in visceral adipose tissue. Continued study of this early post-SG physiology may lead to a better understanding of the anti-diabetic mechanisms of bariatric surgery.


Asunto(s)
Tejido Adiposo/metabolismo , Composición Corporal/fisiología , Gastrectomía , Glucosa/metabolismo , Pérdida de Peso/fisiología , Animales , Glucemia/metabolismo , Calorimetría Indirecta , Ingestión de Alimentos/fisiología , Prueba de Tolerancia a la Glucosa , Homeostasis/fisiología , Masculino , Ratones , Modelos Animales
18.
Hum Mol Genet ; 27(24): 4194-4203, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30169630

RESUMEN

Great strides in gene discovery have been made using a multitude of methods to associate phenotypes with genetic variants, but there still remains a substantial gap between observed symptoms and identified genetic defects. Herein, we use the convergence of various genetic and genomic techniques to investigate the underpinnings of a constellation of phenotypes that include prostate cancer (PCa) and sensorineural hearing loss (SNHL) in a human subject. Through interrogation of the subject's de novo, germline, balanced chromosomal translocation, we first identify a correlation between his disorders and a poorly annotated gene known as lipid droplet associated hydrolase (LDAH). Using data repositories of both germline and somatic variants, we identify convergent genomic evidence that substantiates a correlation between loss of LDAH and PCa. This correlation is validated through both in vitro and in vivo models that show loss of LDAH results in increased risk of PCa and, to a lesser extent, SNHL. By leveraging convergent evidence in emerging genomic data, we hypothesize that loss of LDAH is involved in PCa and other phenotypes observed in support of a genotype-phenotype association in an n-of-one human subject.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Neoplasias de la Próstata/genética , Serina Proteasas/genética , Translocación Genética/genética , Adulto , Anciano , Animales , Estudio de Asociación del Genoma Completo , Células Germinativas/patología , Pérdida Auditiva Sensorineural/patología , Humanos , Masculino , Ratones , Ratones Noqueados , Fenotipo , Neoplasias de la Próstata/patología
19.
Nature ; 508(7495): 258-62, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24717514

RESUMEN

In obesity and type 2 diabetes, Glut4 glucose transporter expression is decreased selectively in adipocytes. Adipose-specific knockout or overexpression of Glut4 alters systemic insulin sensitivity. Here we show, using DNA array analyses, that nicotinamide N-methyltransferase (Nnmt) is the most strongly reciprocally regulated gene when comparing gene expression in white adipose tissue (WAT) from adipose-specific Glut4-knockout or adipose-specific Glut4-overexpressing mice with their respective controls. NNMT methylates nicotinamide (vitamin B3) using S-adenosylmethionine (SAM) as a methyl donor. Nicotinamide is a precursor of NAD(+), an important cofactor linking cellular redox states with energy metabolism. SAM provides propylamine for polyamine biosynthesis and donates a methyl group for histone methylation. Polyamine flux including synthesis, catabolism and excretion, is controlled by the rate-limiting enzymes ornithine decarboxylase (ODC) and spermidine-spermine N(1)-acetyltransferase (SSAT; encoded by Sat1) and by polyamine oxidase (PAO), and has a major role in energy metabolism. We report that NNMT expression is increased in WAT and liver of obese and diabetic mice. Nnmt knockdown in WAT and liver protects against diet-induced obesity by augmenting cellular energy expenditure. NNMT inhibition increases adipose SAM and NAD(+) levels and upregulates ODC and SSAT activity as well as expression, owing to the effects of NNMT on histone H3 lysine 4 methylation in adipose tissue. Direct evidence for increased polyamine flux resulting from NNMT inhibition includes elevated urinary excretion and adipocyte secretion of diacetylspermine, a product of polyamine metabolism. NNMT inhibition in adipocytes increases oxygen consumption in an ODC-, SSAT- and PAO-dependent manner. Thus, NNMT is a novel regulator of histone methylation, polyamine flux and NAD(+)-dependent SIRT1 signalling, and is a unique and attractive target for treating obesity and type 2 diabetes.


Asunto(s)
Dieta , Nicotinamida N-Metiltransferasa/deficiencia , Nicotinamida N-Metiltransferasa/metabolismo , Obesidad/enzimología , Obesidad/prevención & control , Acetiltransferasas/metabolismo , Adipocitos/metabolismo , Tejido Adiposo/enzimología , Tejido Adiposo/metabolismo , Tejido Adiposo Blanco/enzimología , Tejido Adiposo Blanco/metabolismo , Animales , Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético , Hígado Graso , Técnicas de Silenciamiento del Gen , Intolerancia a la Glucosa , Transportador de Glucosa de Tipo 4/deficiencia , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Resistencia a la Insulina , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , NAD/metabolismo , Niacinamida/metabolismo , Nicotinamida N-Metiltransferasa/genética , Obesidad/etiología , Obesidad/genética , Ornitina Descarboxilasa/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , S-Adenosilmetionina/metabolismo , Sirtuina 1/metabolismo , Espermina/análogos & derivados , Espermina/metabolismo , Delgadez/enzimología , Delgadez/metabolismo , Poliamino Oxidasa
20.
Physiol Rev ; 92(3): 1479-514, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22811431

RESUMEN

The sirtuins are a family of highly conserved NAD(+)-dependent deacetylases that act as cellular sensors to detect energy availability and modulate metabolic processes. Two sirtuins that are central to the control of metabolic processes are mammalian sirtuin 1 (SIRT1) and sirtuin 3 (SIRT3), which are localized to the nucleus and mitochondria, respectively. Both are activated by high NAD(+) levels, a condition caused by low cellular energy status. By deacetylating a variety of proteins that induce catabolic processes while inhibiting anabolic processes, SIRT1 and SIRT3 coordinately increase cellular energy stores and ultimately maintain cellular energy homeostasis. Defects in the pathways controlled by SIRT1 and SIRT3 are known to result in various metabolic disorders. Consequently, activation of sirtuins by genetic or pharmacological means can elicit multiple metabolic benefits that protect mice from diet-induced obesity, type 2 diabetes, and nonalcoholic fatty liver disease.


Asunto(s)
Metabolismo Energético , Transducción de Señal , Sirtuina 1/metabolismo , Sirtuina 3/metabolismo , Animales , Ritmo Circadiano , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Activación Enzimática , Activadores de Enzimas/farmacología , Predisposición Genética a la Enfermedad , Homeostasis , Humanos , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Modelos Moleculares , Fenotipo , Polimorfismo Genético , Conformación Proteica , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sirtuina 1/genética , Sirtuina 3/genética , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA