Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Nature ; 604(7905): 337-342, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35355021

RESUMEN

Decades of work have elucidated cytokine signalling and transcriptional pathways that control T cell differentiation and have led the way to targeted biologic therapies that are effective in a range of autoimmune, allergic and inflammatory diseases. Recent evidence indicates that obesity and metabolic disease can also influence the immune system1-7, although the mechanisms and effects on immunotherapy outcomes remain largely unknown. Here, using two models of atopic dermatitis, we show that lean and obese mice mount markedly different immune responses. Obesity converted the classical type 2 T helper (TH2)-predominant disease associated with atopic dermatitis to a more severe disease with prominent TH17 inflammation. We also observed divergent responses to biologic therapies targeting TH2 cytokines, which robustly protected lean mice but exacerbated disease in obese mice. Single-cell RNA sequencing coupled with genome-wide binding analyses revealed decreased activity of nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ) in TH2 cells from obese mice relative to lean mice. Conditional ablation of PPARγ in T cells revealed that PPARγ is required to focus the in vivo TH response towards a TH2-predominant state and prevent aberrant non-TH2 inflammation. Treatment of obese mice with a small-molecule PPARγ agonist limited development of TH17 pathology and unlocked therapeutic responsiveness to targeted anti-TH2 biologic therapies. These studies reveal the effects of obesity on immunological disease and suggest a precision medicine approach to target the immune dysregulation caused by obesity.


Asunto(s)
Dermatitis Atópica , PPAR gamma , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Ratones , Obesidad/metabolismo , PPAR gamma/agonistas , PPAR gamma/metabolismo , Medicina de Precisión , Análisis de Secuencia de ARN , Células Th2/metabolismo
2.
Nat Chem Biol ; 20(2): 221-233, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37884807

RESUMEN

Targeting proximity-labeling enzymes to specific cellular locations is a viable strategy for profiling subcellular proteomes. Here, we generated transgenic mice (MAX-Tg) expressing a mitochondrial matrix-targeted ascorbate peroxidase. Comparative analysis of matrix proteomes from the muscle tissues showed differential enrichment of mitochondrial proteins. We found that reticulon 4-interacting protein 1 (RTN4IP1), also known as optic atrophy-10, is enriched in the mitochondrial matrix of muscle tissues and is an NADPH oxidoreductase. Interactome analysis and in vitro enzymatic assays revealed an essential role for RTN4IP1 in coenzyme Q (CoQ) biosynthesis by regulating the O-methylation activity of COQ3. Rtn4ip1-knockout myoblasts had markedly decreased CoQ9 levels and impaired cellular respiration. Furthermore, muscle-specific knockdown of dRtn4ip1 in flies resulted in impaired muscle function, which was reversed by dietary supplementation with soluble CoQ. Collectively, these results demonstrate that RTN4IP1 is a mitochondrial NAD(P)H oxidoreductase essential for supporting mitochondrial respiration activity in the muscle tissue.


Asunto(s)
Oxidorreductasas , Ubiquinona , Animales , Ratones , Drosophila melanogaster , Ratones Transgénicos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Proteoma , Ubiquinona/metabolismo , Proteínas Portadoras
3.
Proc Natl Acad Sci U S A ; 120(20): e2219644120, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37155882

RESUMEN

Emerging evidence suggest that transcription factors play multiple roles in the development of pancreatitis, a necroinflammatory condition lacking specific therapy. Estrogen-related receptor γ (ERRγ), a pleiotropic transcription factor, has been reported to play a vital role in pancreatic acinar cell (PAC) homeostasis. However, the role of ERRγ in PAC dysfunction remains hitherto unknown. Here, we demonstrated in both mice models and human cohorts that pancreatitis is associated with an increase in ERRγ gene expression via activation of STAT3. Acinar-specific ERRγ haploinsufficiency or pharmacological inhibition of ERRγ significantly impaired the progression of pancreatitis both in vitro and in vivo. Using systematic transcriptomic analysis, we identified that voltage-dependent anion channel 1 (VDAC1) acts as a molecular mediator of ERRγ. Mechanistically, we showed that induction of ERRγ in cultured acinar cells and mouse pancreata enhanced VDAC1 expression by directly binding to specific site of the Vdac1 gene promoter and resulted in VDAC1 oligomerization. Notably, VDAC1, whose expression and oligomerization were dependent on ERRγ, modulates mitochondrial Ca2+ and ROS levels. Inhibition of the ERRγ-VDAC1 axis could alleviate mitochondrial Ca2+ accumulation, ROS formation and inhibit progression of pancreatitis. Using two different mouse models of pancreatitis, we showed that pharmacological blockade of ERRγ-VDAC1 pathway has therapeutic benefits in mitigating progression of pancreatitis. Likewise, using PRSS1R122H-Tg mice to mimic human hereditary pancreatitis, we demonstrated that ERRγ inhibitor also alleviated pancreatitis. Our findings highlight the importance of ERRγ in pancreatitis progression and suggests its therapeutic intervention for prevention and treatment of pancreatitis.


Asunto(s)
Pancreatitis Crónica , Canal Aniónico 1 Dependiente del Voltaje , Animales , Humanos , Ratones , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba , Canal Aniónico 1 Dependiente del Voltaje/metabolismo
4.
Mol Ther ; 31(4): 1002-1016, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-36755495

RESUMEN

Fabry disease (FD), a lysosomal storage disorder, is caused by defective α-galactosidase (GLA) activity, which results in the accumulation of globotriaosylceramide (Gb3) in endothelial cells and leads to life-threatening complications such as left ventricular hypertrophy (LVH), renal failure, and stroke. Enzyme replacement therapy (ERT) results in Gb3 clearance; however, because of a short half-life in the body and the high immunogenicity of FD patients, ERT has a limited therapeutic effect, particularly in patients with late-onset disease or progressive complications. Because vascular endothelial cells (VECs) derived from FD-induced pluripotent stem cells display increased thrombospondin-1 (TSP1) expression and enhanced SMAD2 signaling, we screened for chemical compounds that could downregulate TSP1 and SMAD2 signaling. Fasudil reduced the levels of p-SMAD2 and TSP1 in FD-VECs and increased the expression of angiogenic factors. Furthermore, fasudil downregulated the endothelial-to-mesenchymal transition (EndMT) and mitochondrial function of FD-VECs. Oral administration of fasudil to FD mice alleviated several FD phenotypes, including LVH, renal fibrosis, anhidrosis, and heat insensitivity. Our findings demonstrate that fasudil is a novel candidate for FD therapy.


Asunto(s)
Enfermedad de Fabry , Animales , Ratones , Enfermedad de Fabry/tratamiento farmacológico , Enfermedad de Fabry/genética , Células Endoteliales/metabolismo , alfa-Galactosidasa/genética , Fenotipo , Terapia de Reemplazo Enzimático
5.
Gastroenterology ; 163(1): 239-256, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35461826

RESUMEN

BACKGROUND & AIMS: Mitochondrial dysfunction disrupts the synthesis and secretion of digestive enzymes in pancreatic acinar cells and plays a primary role in the etiology of exocrine pancreas disorders. However, the transcriptional mechanisms that regulate mitochondrial function to support acinar cell physiology are poorly understood. Here, we aim to elucidate the function of estrogen-related receptor γ (ERRγ) in pancreatic acinar cell mitochondrial homeostasis and energy production. METHODS: Two models of ERRγ inhibition, GSK5182-treated wild-type mice and ERRγ conditional knock-out (cKO) mice, were established to investigate ERRγ function in the exocrine pancreas. To identify the functional role of ERRγ in pancreatic acinar cells, we performed histologic and transcriptome analysis with the pancreas isolated from ERRγ cKO mice. To determine the relevance of these findings for human disease, we analyzed transcriptome data from multiple independent human cohorts and conducted genetic association studies for ESRRG variants in 2 distinct human pancreatitis cohorts. RESULTS: Blocking ERRγ function in mice by genetic deletion or inverse agonist treatment results in striking pancreatitis-like phenotypes accompanied by inflammation, fibrosis, and cell death. Mechanistically, loss of ERRγ in primary acini abrogates messenger RNA expression and protein levels of mitochondrial oxidative phosphorylation complex genes, resulting in defective acinar cell energetics. Mitochondrial dysfunction due to ERRγ deletion further triggers autophagy dysfunction, endoplasmic reticulum stress, and production of reactive oxygen species, ultimately leading to cell death. Interestingly, ERRγ-deficient acinar cells that escape cell death acquire ductal cell characteristics, indicating a role for ERRγ in acinar-to-ductal metaplasia. Consistent with our findings in ERRγ cKO mice, ERRγ expression was significantly reduced in patients with chronic pancreatitis compared with normal subjects. Furthermore, candidate locus region genetic association studies revealed multiple single nucleotide variants for ERRγ that are associated with chronic pancreatitis. CONCLUSIONS: Collectively, our findings highlight an essential role for ERRγ in maintaining the transcriptional program that supports acinar cell mitochondrial function and organellar homeostasis and provide a novel molecular link between ERRγ and exocrine pancreas disorders.


Asunto(s)
Páncreas Exocrino , Pancreatitis Crónica , Células Acinares/patología , Animales , Estrógenos/metabolismo , Humanos , Ratones , Ratones Noqueados , Páncreas/patología , Páncreas Exocrino/metabolismo , Pancreatitis Crónica/patología
6.
Molecules ; 27(11)2022 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-35684355

RESUMEN

Serotonin (5-hydroxytryptophan) is a hormone that regulates emotions in the central nervous system. However, serotonin in the peripheral system is associated with obesity and fatty liver disease. Because serotonin cannot cross the blood-brain barrier (BBB), we focused on identifying new tryptophan hydroxylase type I (TPH1) inhibitors that act only in peripheral tissues for treating obesity and fatty liver disease without affecting the central nervous system. Structural optimization inspired by para-chlorophenylalanine (pCPA) resulted in the identification of a series of oxyphenylalanine and heterocyclic phenylalanine derivatives as TPH1 inhibitors. Among these compounds, compound 18i with an IC50 value of 37 nM was the most active in vitro. Additionally, compound 18i showed good liver microsomal stability and did not significantly inhibit CYP and Herg. Furthermore, this TPH1 inhibitor was able to actively interact with the peripheral system without penetrating the BBB. Compound 18i and its prodrug reduced body weight gain in mammals and decreased in vivo fat accumulation.


Asunto(s)
Hepatopatías , Triptófano Hidroxilasa , Animales , Barrera Hematoencefálica/metabolismo , Mamíferos/metabolismo , Obesidad/tratamiento farmacológico , Serotonina , Triptófano Hidroxilasa/metabolismo
7.
Hepatology ; 71(3): 1055-1069, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31355949

RESUMEN

BACKGROUND AND AIMS: Liver receptor homolog-1 (LRH-1; NR5A2) is a nuclear receptor that regulates metabolic homeostasis in the liver. Previous studies identified phosphatidylcholines as potential endogenous agonist ligands for LRH-1. In the liver, distinct subsets of phosphatidylcholine species are generated by two different pathways: choline addition to phosphatidic acid through the Kennedy pathway and trimethylation of phosphatidylethanolamine through phosphatidylethanolamine N-methyl transferase (PEMT). APPROACH AND RESULTS: Here, we report that a PEMT-LRH-1 pathway specifically couples methyl metabolism and mitochondrial activities in hepatocytes. We show that the loss of Lrh-1 reduces mitochondrial number, basal respiration, beta-oxidation, and adenosine triphosphate production in hepatocytes and decreases expression of mitochondrial biogenesis and beta-oxidation genes. In contrast, activation of LRH-1 by its phosphatidylcholine agonists exerts opposite effects. While disruption of the Kennedy pathway does not affect the LRH-1-mediated regulation of mitochondrial activities, genetic or pharmaceutical inhibition of the PEMT pathway recapitulates the effects of Lrh-1 knockdown on mitochondria. Furthermore, we show that S-adenosyl methionine, a cofactor required for PEMT, is sufficient to induce Lrh-1 transactivation and consequently mitochondrial biogenesis. CONCLUSIONS: A PEMT-LRH-1 axis regulates mitochondrial biogenesis and beta-oxidation in hepatocytes.


Asunto(s)
Hepatocitos/metabolismo , Mitocondrias/fisiología , Fosfatidiletanolamina N-Metiltransferasa/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Células Hep G2 , Humanos , Masculino , Ratones , Oxidación-Reducción , S-Adenosilmetionina/metabolismo , S-Adenosilmetionina/farmacología
8.
Nature ; 513(7518): 436-9, 2014 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-25043058

RESUMEN

Fibroblast growth factor 1 (FGF1) is an autocrine/paracrine regulator whose binding to heparan sulphate proteoglycans effectively precludes its circulation. Although FGF1 is known as a mitogenic factor, FGF1 knockout mice develop insulin resistance when stressed by a high-fat diet, suggesting a potential role in nutrient homeostasis. Here we show that parenteral delivery of a single dose of recombinant FGF1 (rFGF1) results in potent, insulin-dependent lowering of glucose levels in diabetic mice that is dose-dependent but does not lead to hypoglycaemia. Chronic pharmacological treatment with rFGF1 increases insulin-dependent glucose uptake in skeletal muscle and suppresses the hepatic production of glucose to achieve whole-body insulin sensitization. The sustained glucose lowering and insulin sensitization attributed to rFGF1 are not accompanied by the side effects of weight gain, liver steatosis and bone loss associated with current insulin-sensitizing therapies. We also show that the glucose-lowering activity of FGF1 can be dissociated from its mitogenic activity and is mediated predominantly via FGF receptor 1 signalling. Thus we have uncovered an unexpected, neomorphic insulin-sensitizing action for exogenous non-mitogenic human FGF1 with therapeutic potential for the treatment of insulin resistance and type 2 diabetes.


Asunto(s)
Factor 1 de Crecimiento de Fibroblastos/farmacología , Glucosa/metabolismo , Insulina/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Relación Dosis-Respuesta a Droga , Factor 1 de Crecimiento de Fibroblastos/administración & dosificación , Factor 1 de Crecimiento de Fibroblastos/efectos adversos , Prueba de Tolerancia a la Glucosa , Humanos , Resistencia a la Insulina , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Mitógenos/farmacología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
9.
Nature ; 485(7398): 391-4, 2012 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-22522926

RESUMEN

Although feast and famine cycles illustrate that remodelling of adipose tissue in response to fluctuations in nutrient availability is essential for maintaining metabolic homeostasis, the underlying mechanisms remain poorly understood. Here we identify fibroblast growth factor 1 (FGF1) as a critical transducer in this process in mice, and link its regulation to the nuclear receptor PPARγ (peroxisome proliferator activated receptor γ), which is the adipocyte master regulator and the target of the thiazolidinedione class of insulin sensitizing drugs. FGF1 is the prototype of the 22-member FGF family of proteins and has been implicated in a range of physiological processes, including development, wound healing and cardiovascular changes. Surprisingly, FGF1 knockout mice display no significant phenotype under standard laboratory conditions. We show that FGF1 is highly induced in adipose tissue in response to a high-fat diet and that mice lacking FGF1 develop an aggressive diabetic phenotype coupled to aberrant adipose expansion when challenged with a high-fat diet. Further analysis of adipose depots in FGF1-deficient mice revealed multiple histopathologies in the vasculature network, an accentuated inflammatory response, aberrant adipocyte size distribution and ectopic expression of pancreatic lipases. On withdrawal of the high-fat diet, this inflamed adipose tissue fails to properly resolve, resulting in extensive fat necrosis. In terms of mechanisms, we show that adipose induction of FGF1 in the fed state is regulated by PPARγ acting through an evolutionarily conserved promoter proximal PPAR response element within the FGF1 gene. The discovery of a phenotype for the FGF1 knockout mouse establishes the PPARγ­FGF1 axis as critical for maintaining metabolic homeostasis and insulin sensitization.


Asunto(s)
Factor 1 de Crecimiento de Fibroblastos/genética , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Homeostasis , Grasa Intraabdominal/metabolismo , PPAR gamma/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos/patología , Animales , Secuencia de Bases , Tamaño de la Célula/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Dieta Alta en Grasa/efectos adversos , Factor 1 de Crecimiento de Fibroblastos/deficiencia , Homeostasis/efectos de los fármacos , Humanos , Inflamación/genética , Insulina/metabolismo , Resistencia a la Insulina , Grasa Intraabdominal/efectos de los fármacos , Grasa Intraabdominal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis/enzimología , Regiones Promotoras Genéticas/genética , Elementos de Respuesta/genética
11.
Proc Natl Acad Sci U S A ; 108(8): 3412-7, 2011 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-21300871

RESUMEN

The ligand-dependent competing actions of nuclear receptor (NR)-associated transcriptional corepressor and coactivator complexes allow for the precise regulation of NR-dependent gene expression in response to both temporal and environmental cues. Here we report the mouse model termed silencing mediator of retinoid and thyroid hormone receptors (SMRT)(mRID1) in which targeted disruption of the first receptor interaction domain (RID) of the nuclear corepressor SMRT disrupts interactions with a subset of NRs and leads to diet-induced superobesity associated with a depressed respiratory exchange ratio, decreased ambulatory activity, and insulin resistance. Although apparently normal when chow fed, SMRT(mRID1) mice develop multiple metabolic dysfunctions when challenged by a high-fat diet, manifested by marked lipid accumulation in white and brown adipose tissue and the liver. The increased weight gain of SMRT(mRID1) mice on a high-fat diet occurs predominantly in fat with adipocyte hypertrophy evident in both visceral and s.c. depots. Importantly, increased inflammatory gene expression was detected only in the visceral depots. SMRT(mRID1) mice are both insulin-insensitive and refractory to the glucose-lowering effects of TZD and AICAR. Increased serum cholesterol and triglyceride levels were observed, accompanied by increased leptin and decreased adiponectin levels. Aberrant storage of lipids in the liver occurred as triglycerides and cholesterol significantly compromised hepatic function. Lipid accumulation in brown adipose tissue was associated with reduced thermogenic capacity and mitochondrial biogenesis. Collectively, these studies highlight the essential role of NR corepressors in maintaining metabolic homeostasis and describe an essential role for SMRT in regulating the progression, severity, and therapeutic outcome of metabolic diseases.


Asunto(s)
Tejido Adiposo/metabolismo , Dieta/efectos adversos , Resistencia a la Insulina , Co-Represor 2 de Receptor Nuclear/fisiología , Obesidad/etiología , Fosforilación Oxidativa , Animales , Homeostasis , Metabolismo de los Lípidos , Hígado/metabolismo , Ratones
12.
Nat Metab ; 6(5): 847-860, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38811804

RESUMEN

Adipose tissues serve as an energy reservoir and endocrine organ, yet the mechanisms that coordinate these functions remain elusive. Here, we show that the transcriptional coregulators, YAP and TAZ, uncouple fat mass from leptin levels and regulate adipocyte plasticity to maintain metabolic homeostasis. Activating YAP/TAZ signalling in adipocytes by deletion of the upstream regulators Lats1 and Lats2 results in a profound reduction in fat mass by converting mature adipocytes into delipidated progenitor-like cells, but does not cause lipodystrophy-related metabolic dysfunction, due to a paradoxical increase in circulating leptin levels. Mechanistically, we demonstrate that YAP/TAZ-TEAD signalling upregulates leptin expression by directly binding to an upstream enhancer site of the leptin gene. We further show that YAP/TAZ activity is associated with, and functionally required for, leptin regulation during fasting and refeeding. These results suggest that adipocyte Hippo-YAP/TAZ signalling constitutes a nexus for coordinating adipose tissue lipid storage capacity and systemic energy balance through the regulation of adipocyte plasticity and leptin gene transcription.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Adipocitos , Tejido Adiposo , Metabolismo Energético , Vía de Señalización Hippo , Leptina , Proteínas Serina-Treonina Quinasas , Transducción de Señal , Proteínas Señalizadoras YAP , Animales , Leptina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Señalizadoras YAP/metabolismo , Tejido Adiposo/metabolismo , Adipocitos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Transactivadores/metabolismo , Transactivadores/genética
13.
Cell Rep ; 42(6): 112636, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37310859

RESUMEN

Obesity-mediated hypoxic stress underlies inflammation, including interferon (IFN)-γ production by natural killer (NK) cells in white adipose tissue. However, the effects of obesity on NK cell IFN-γ production remain obscure. Here, we show that hypoxia promotes xCT-mediated glutamate excretion and C-X-C motif chemokine ligand 12 (CXCL12) expression in white adipocytes, resulting in CXCR4+ NK cell recruitment. Interestingly, this spatial proximity between adipocytes and NK cells induces IFN-γ production in NK cells by stimulating metabotropic glutamate receptor 5 (mGluR5). IFN-γ then triggers inflammatory activation of macrophages and augments xCT and CXCL12 expression in adipocytes, forming a bidirectional pathway. Genetic or pharmacological inhibition of xCT, mGluR5, or IFN-γ receptor in adipocytes or NK cells alleviates obesity-related metabolic disorders in mice. Consistently, patients with obesity showed elevated levels of glutamate/mGluR5 and CXCL12/CXCR4 axes, suggesting that a bidirectional pathway between adipocytes and NK cells could be a viable therapeutic target in obesity-related metabolic disorders.


Asunto(s)
Adipocitos Blancos , Ácido Glutámico , Interferón gamma , Obesidad , Animales , Humanos , Ratones , Adipocitos Blancos/metabolismo , Ácido Glutámico/metabolismo , Interferón gamma/metabolismo , Células Asesinas Naturales/metabolismo , Obesidad/metabolismo , Sistema de Transporte de Aminoácidos y+/metabolismo
14.
Nat Commun ; 14(1): 3746, 2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37353518

RESUMEN

Brown adipose tissue (BAT) has abundant mitochondria with the unique capability of generating heat via uncoupled respiration. Mitochondrial uncoupling protein 1 (UCP1) is activated in BAT during cold stress and dissipates mitochondrial proton motive force generated by the electron transport chain to generate heat. However, other mitochondrial factors required for brown adipocyte respiration and thermogenesis under cold stress are largely unknown. Here, we show LETM1 domain-containing protein 1 (LETMD1) is a BAT-enriched and cold-induced protein required for cold-stimulated respiration and thermogenesis of BAT. Proximity labeling studies reveal that LETMD1 is a mitochondrial matrix protein. Letmd1 knockout male mice display aberrant BAT mitochondria and fail to carry out adaptive thermogenesis under cold stress. Letmd1 knockout BAT is deficient in oxidative phosphorylation (OXPHOS) complex proteins and has impaired mitochondrial respiration. In addition, BAT-specific Letmd1 deficient mice exhibit phenotypes identical to those observed in Letmd1 knockout mice. Collectively, we demonstrate that the BAT-enriched mitochondrial matrix protein LETMD1 plays a tissue-autonomous role that is essential for BAT mitochondrial function and thermogenesis.


Asunto(s)
Tejido Adiposo Pardo , Proteínas Mitocondriales , Termogénesis , Animales , Masculino , Ratones , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/metabolismo , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Termogénesis/genética , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
15.
Cell Metab ; 6(3): 195-207, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17767906

RESUMEN

Adipose (Adp) is an evolutionarily conserved gene isolated from naturally occurring obese flies homozygous for an adp mutation. Here we show that the anti-obesity function of Adp (worm Y73E7A.9, fly adp, and murine Wdtc1) is conserved from worms to mammals. Further, Adp appears to inhibit fat formation in a dosage-sensitive manner. Adp heterozygous flies and Adp heterozygous mutant mice are obese and insulin resistant, as are mice that express a dominant negative form of Adp in fat cells. Conversely, fat-restricted Adp transgenic mice are lean and display improved metabolic profiles. A transient transgenic increase in Adp activity in adult fly fat tissues reduces fat accumulation, indicating therapeutic potential. ADP may elicit these anti-adipogenic functions by regulating chromatin dynamics and gene transcription, as it binds both histones and HDAC3 and inhibits PPARgamma activity. Thus Adp appears to be involved in an ancient pathway that regulates fat accumulation.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Obesidad/genética , Proteínas/genética , Proteínas/metabolismo , Tejido Adiposo/anatomía & histología , Tejido Adiposo/fisiología , Animales , Caenorhabditis elegans/anatomía & histología , Caenorhabditis elegans/fisiología , Proteínas de Caenorhabditis elegans/antagonistas & inhibidores , Proteínas de Drosophila/antagonistas & inhibidores , Femenino , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Hiperglucemia/metabolismo , Hiperglucemia/prevención & control , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Obesidad/prevención & control , Proteínas/antagonistas & inhibidores
16.
Lab Chip ; 22(20): 3920-3932, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36097851

RESUMEN

Adipocyte dedifferentiation has recently gained attention as a process underpinning adipocyte plasticity; however, a lack of suitable experimental platforms has hampered studies into the underlying mechanisms. Here, we developed a microscope-mountable ceiling culture chip that provides a stable yet tunable culture environment for long-term live-imaging of dedifferentiating adipocytes. A detailed spatiotemporal analysis of mature adipocyte dedifferentiation utilizing the culture platform and Cre-recombinase tracers revealed the involvement of dynamic actin remodeling for lipid droplet (LD) secretion during adipocyte dedifferentiation. Additionally, Hippo, Hedgehog, and PPARγ signaling pathways were identified as potent regulators of adipocyte dedifferentiation. Contrary to the belief that adult adipocytes are relatively static, we show that adipocytes are very dynamic, relying on actin-driven mechanical forces to execute LD extrusion and intercellular LD transfer processes.


Asunto(s)
Actinas , Gotas Lipídicas , Adipocitos/metabolismo , Desdiferenciación Celular , Gotas Lipídicas/metabolismo , PPAR gamma/metabolismo , Recombinasas/metabolismo
17.
Cell Metab ; 3(1): 25-34, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16399502

RESUMEN

Hedgehog (Hh) signals regulate invertebrate and vertebrate development, yet the role of the cascade in adipose development was undefined. To analyze a potential function, we turned to Drosophila and mammalian models. Fat-body-specific transgenic activation of Hh signaling inhibits fly fat formation. Conversely, fat-body-specific Hh blockade stimulated fly fat formation. In mammalian models, sufficiency and necessity tests showed that Hh signaling also inhibits mammalian adipogenesis. Hh signals elicit this function early in adipogenesis, upstream of PPARgamma, potentially diverting preadipocytes as well as multipotent mesenchymal prescursors away from adipogenesis and toward osteogenesis. Hh may elicit these effects by inducing the expression of antiadipogenic transcription factors such as Gata2. These data support the notion that Hh signaling plays a conserved role, from invertebrates to vertebrates, in inhibiting fat formation and highlighting the potential of the Hh pathway as a therapeutic target for osteoporosis, lipodystrophy, diabetes, and obesity.


Asunto(s)
Adipogénesis/fisiología , Secuencia Conservada , Proteínas de Drosophila/fisiología , Proteínas Hedgehog/fisiología , Transducción de Señal/fisiología , Células 3T3-L1 , Tejido Adiposo/fisiología , Animales , Biomarcadores , Modelos Animales de Enfermedad , Proteínas de Drosophila/agonistas , Drosophila melanogaster/fisiología , Evolución Molecular , Cuerpo Adiposo/fisiología , Factores de Transcripción GATA/fisiología , Proteínas Hedgehog/agonistas , Ratones , Ratones Endogámicos C3H , Células Madre Multipotentes/fisiología , Células 3T3 NIH , Obesidad/metabolismo , Osteogénesis/fisiología , PPAR gamma/fisiología
18.
Nat Commun ; 12(1): 5204, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34471136

RESUMEN

Secretory proteins are an essential component of interorgan communication networks that regulate animal physiology. Current approaches for identifying secretory proteins from specific cell and tissue types are largely limited to in vitro or ex vivo models which often fail to recapitulate in vivo biology. As such, there is mounting interest in developing in vivo analytical tools that can provide accurate information on the origin, identity, and spatiotemporal dynamics of secretory proteins. Here, we describe iSLET (in situ Secretory protein Labeling via ER-anchored TurboID) which selectively labels proteins that transit through the classical secretory pathway via catalytic actions of Sec61b-TurboID, a proximity labeling enzyme anchored in the ER lumen. To validate iSLET in a whole-body system, we express iSLET in the mouse liver and demonstrate efficient labeling of liver secretory proteins which could be tracked and identified within circulating blood plasma. Furthermore, proteomic analysis of the labeled liver secretome enriched from liver iSLET mouse plasma is highly consistent with previous reports of liver secretory protein profiles. Taken together, iSLET is a versatile and powerful tool for studying spatiotemporal dynamics of secretory proteins, a valuable class of biomarkers and therapeutic targets.


Asunto(s)
Retículo Endoplásmico/metabolismo , Canales de Translocación SEC/metabolismo , Vías Secretoras/fisiología , Animales , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Proteoma/metabolismo , Proteómica
19.
Nat Cell Biol ; 23(2): 172-183, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33558728

RESUMEN

In patients with advanced-stage cancer, cancer-associated anorexia affects treatment success and patient survival. However, the underlying mechanism is poorly understood. Here, we show that Dilp8, a Drosophila homologue of mammalian insulin-like 3 peptide (INSL3), is secreted from tumour tissues and induces anorexia through the Lgr3 receptor in the brain. Activated Dilp8-Lgr3 signalling upregulated anorexigenic nucleobinding 1 (NUCB1) and downregulated orexigenic short neuropeptide F (sNPF) and NPF expression in the brain. In the cancer condition, the protein expression of Lgr3 and NUCB1 was significantly upregulated in neurons expressing sNPF and NPF. INSL3 levels were increased in tumour-implanted mice and INSL3-treated mouse hypothalamic cells showed Nucb2 upregulation and Npy downregulation. Food consumption was significantly reduced in intracerebrospinal INSL3-injected mice. In patients with pancreatic cancer, higher serum INSL3 levels increased anorexia. These results indicate that tumour-derived Dilp8/INSL3 induces cancer anorexia by regulating feeding hormones through the Lgr3/Lgr8 receptor in Drosophila and mammals.


Asunto(s)
Anorexia/metabolismo , Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias/metabolismo , Neuropéptidos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Secuencia de Aminoácidos , Animales , Anorexia/etiología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Neoplasias del Ojo/patología , Conducta Alimentaria , Humanos , Hipotálamo/metabolismo , Insulina/sangre , Insulina/química , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones Endogámicos C57BL , Neoplasias/complicaciones , Neuronas/metabolismo , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/complicaciones , Proteínas/química , Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
20.
J Clin Invest ; 131(23)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34618686

RESUMEN

Insulin resistance is a cornerstone of obesity-related complications such as type 2 diabetes, metabolic syndrome, and nonalcoholic fatty liver disease. A high rate of lipolysis is known to be associated with insulin resistance, and inhibiting adipose tissue lipolysis improves obesity-related insulin resistance. Here, we demonstrate that inhibition of serotonin (5-hydroxytryptamine [5-HT]) signaling through serotonin receptor 2B (HTR2B) in adipose tissues ameliorates insulin resistance by reducing lipolysis in visceral adipocytes. Chronic high-fat diet (HFD) feeding increased Htr2b expression in epididymal white adipose tissue, resulting in increased HTR2B signaling in visceral white adipose tissue. Moreover, HTR2B expression in white adipose tissue was increased in obese humans and positively correlated with metabolic parameters. We further found that adipocyte-specific Htr2b-knockout mice are resistant to HFD-induced insulin resistance, visceral adipose tissue inflammation, and hepatic steatosis. Enhanced 5-HT signaling through HTR2B directly activated lipolysis through phosphorylation of hormone-sensitive lipase in visceral adipocytes. Moreover, treatment with a selective HTR2B antagonist attenuated HFD-induced insulin resistance, visceral adipose tissue inflammation, and hepatic steatosis. Thus, adipose HTR2B signaling could be a potential therapeutic target for treatment of obesity-related insulin resistance.


Asunto(s)
Resistencia a la Insulina , Grasa Intraabdominal/metabolismo , Obesidad/metabolismo , Receptor de Serotonina 5-HT2B/metabolismo , Serotonina/metabolismo , Adipocitos/citología , Adipocitos Blancos , Tejido Adiposo , Tejido Adiposo Blanco/metabolismo , Adulto , Animales , Dieta Alta en Grasa , Epidídimo , Femenino , Glicerol/metabolismo , Humanos , Inflamación , Insulina/metabolismo , Lipólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Fosforilación , Transducción de Señal , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA