Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell ; 181(6): 1246-1262.e22, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32442405

RESUMEN

There is considerable inter-individual variability in susceptibility to weight gain despite an equally obesogenic environment in large parts of the world. Whereas many studies have focused on identifying the genetic susceptibility to obesity, we performed a GWAS on metabolically healthy thin individuals (lowest 6th percentile of the population-wide BMI spectrum) in a uniquely phenotyped Estonian cohort. We discovered anaplastic lymphoma kinase (ALK) as a candidate thinness gene. In Drosophila, RNAi mediated knockdown of Alk led to decreased triglyceride levels. In mice, genetic deletion of Alk resulted in thin animals with marked resistance to diet- and leptin-mutation-induced obesity. Mechanistically, we found that ALK expression in hypothalamic neurons controls energy expenditure via sympathetic control of adipose tissue lipolysis. Our genetic and mechanistic experiments identify ALK as a thinness gene, which is involved in the resistance to weight gain.


Asunto(s)
Quinasa de Linfoma Anaplásico/genética , Delgadez/genética , Tejido Adiposo/metabolismo , Adulto , Animales , Línea Celular , Estudios de Cohortes , Drosophila/genética , Estonia , Femenino , Humanos , Leptina/genética , Lipólisis/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Interferencia de ARN/fisiología , Adulto Joven
2.
Cell ; 164(3): 353-64, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-26824653

RESUMEN

More than one-half billion people are obese, and despite progress in genetic research, much of the heritability of obesity remains enigmatic. Here, we identify a Trim28-dependent network capable of triggering obesity in a non-Mendelian, "on/off" manner. Trim28(+/D9) mutant mice exhibit a bi-modal body-weight distribution, with isogenic animals randomly emerging as either normal or obese and few intermediates. We find that the obese-"on" state is characterized by reduced expression of an imprinted gene network including Nnat, Peg3, Cdkn1c, and Plagl1 and that independent targeting of these alleles recapitulates the stochastic bi-stable disease phenotype. Adipose tissue transcriptome analyses in children indicate that humans too cluster into distinct sub-populations, stratifying according to Trim28 expression, transcriptome organization, and obesity-associated imprinted gene dysregulation. These data provide evidence of discrete polyphenism in mouse and man and thus carry important implications for complex trait genetics, evolution, and medicine.


Asunto(s)
Epigénesis Genética , Haploinsuficiencia , Proteínas Nucleares/genética , Obesidad/genética , Proteínas Represoras/genética , Delgadez/genética , Adolescente , Animales , Índice de Masa Corporal , Niño , Preescolar , Humanos , Ratones , Encuestas Nutricionales , Polimorfismo Genético , Proteína 28 que Contiene Motivos Tripartito
3.
Cell ; 140(1): 148-60, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-20074523

RESUMEN

Over 1 billion people are estimated to be overweight, placing them at risk for diabetes, cardiovascular disease, and cancer. We performed a systems-level genetic dissection of adiposity regulation using genome-wide RNAi screening in adult Drosophila. As a follow-up, the resulting approximately 500 candidate obesity genes were functionally classified using muscle-, oenocyte-, fat-body-, and neuronal-specific knockdown in vivo and revealed hedgehog signaling as the top-scoring fat-body-specific pathway. To extrapolate these findings into mammals, we generated fat-specific hedgehog-activation mutant mice. Intriguingly, these mice displayed near total loss of white, but not brown, fat compartments. Mechanistically, activation of hedgehog signaling irreversibly blocked differentiation of white adipocytes through direct, coordinate modulation of early adipogenic factors. These findings identify a role for hedgehog signaling in white/brown adipocyte determination and link in vivo RNAi-based scanning of the Drosophila genome to regulation of adipocyte cell fate in mammals.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/metabolismo , Obesidad/genética , Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Adipogénesis , Animales , AMP Cíclico/metabolismo , Glucocorticoides/metabolismo , Humanos , Ratones , Ratones Noqueados , Células Musculares/metabolismo , Proteínas Represoras/genética
4.
Genes Dev ; 31(20): 2099-2112, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-29118048

RESUMEN

Lung cancer is the leading cause of cancer deaths. Besides smoking, epidemiological studies have linked female sex hormones to lung cancer in women; however, the underlying mechanisms remain unclear. Here we report that the receptor activator of nuclear factor-kB (RANK), the key regulator of osteoclastogenesis, is frequently expressed in primary lung tumors, an active RANK pathway correlates with decreased survival, and pharmacologic RANK inhibition reduces tumor growth in patient-derived lung cancer xenografts. Clonal genetic inactivation of KRasG12D in mouse lung epithelial cells markedly impairs the progression of KRasG12D -driven lung cancer, resulting in a significant survival advantage. Mechanistically, RANK rewires energy homeostasis in human and murine lung cancer cells and promotes expansion of lung cancer stem-like cells, which is blocked by inhibiting mitochondrial respiration. Our data also indicate survival differences in KRasG12D -driven lung cancer between male and female mice, and we show that female sex hormones can promote lung cancer progression via the RANK pathway. These data uncover a direct role for RANK in lung cancer and may explain why female sex hormones accelerate lung cancer development. Inhibition of RANK using the approved drug denosumab may be a therapeutic drug candidate for primary lung cancer.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Receptor Activador del Factor Nuclear kappa-B/fisiología , Células Epiteliales Alveolares/metabolismo , Animales , Respiración de la Célula , Células Cultivadas , Metabolismo Energético , Femenino , Hormonas Esteroides Gonadales/fisiología , Homeostasis , Humanos , Pulmón/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Masculino , Ratones , Mitocondrias/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptor Activador del Factor Nuclear kappa-B/antagonistas & inhibidores , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Mucosa Respiratoria/metabolismo
5.
Nature ; 495(7442): 474-80, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23474986

RESUMEN

CLP1 was the first mammalian RNA kinase to be identified. However, determining its in vivo function has been elusive. Here we generated kinase-dead Clp1 (Clp1(K/K)) mice that show a progressive loss of spinal motor neurons associated with axonal degeneration in the peripheral nerves and denervation of neuromuscular junctions, resulting in impaired motor function, muscle weakness, paralysis and fatal respiratory failure. Transgenic rescue experiments show that CLP1 functions in motor neurons. Mechanistically, loss of CLP1 activity results in accumulation of a novel set of small RNA fragments, derived from aberrant processing of tyrosine pre-transfer RNA. These tRNA fragments sensitize cells to oxidative-stress-induced p53 (also known as TRP53) activation and p53-dependent cell death. Genetic inactivation of p53 rescues Clp1(K/K) mice from the motor neuron loss, muscle denervation and respiratory failure. Our experiments uncover a mechanistic link between tRNA processing, formation of a new RNA species and progressive loss of lower motor neurons regulated by p53.


Asunto(s)
Neuronas Motoras/metabolismo , Neuronas Motoras/patología , ARN de Transferencia de Tirosina/metabolismo , Factores de Transcripción/metabolismo , Esclerosis Amiotrófica Lateral , Animales , Animales Recién Nacidos , Axones/metabolismo , Axones/patología , Muerte Celular , Diafragma/inervación , Pérdida del Embrión , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Exones/genética , Femenino , Fibroblastos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Atrofia Muscular Espinal , Enfermedades Neuromusculares/metabolismo , Enfermedades Neuromusculares/patología , Estrés Oxidativo , Procesamiento Postranscripcional del ARN , ARN de Transferencia de Tirosina/genética , Proteínas de Unión al ARN , Respiración , Nervios Espinales/citología , Factores de Transcripción/deficiencia , Proteína p53 Supresora de Tumor/metabolismo , Tirosina/genética , Tirosina/metabolismo
6.
Eur J Clin Invest ; 46(11): 911-919, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27600500

RESUMEN

BACKGROUND: Infiltration of white adipose tissue (WAT) by inflammatory cells in obesity is considered to be a key event in the development of insulin resistance. Recently, mast cells (MCs) have been identified as new players in the pathogenesis of obesity. We aimed to investigate the relationship between MCs and various inflammatory markers in serum and WAT and to determine the role of MCs in the aetiology of insulin resistance. MATERIALS AND METHODS: Gene expression was measured in WAT from 20 morbidly obese patients and 20 nonobese control subjects. Homoeostasis Model of Assessment-Insulin Resistance (HOMA-IR) was used to estimate insulin sensitivity. In addition, wild-type and mast cell-deficient mice were fed a high-fat or low-fat diet to study mast cell influence on inflammatory cell polarization in WAT and overall metabolic changes. RESULTS: WAT levels of MC-specific TPSb2 transcript were increased in obesity and significantly positively correlated with TNF, CCL2, CCL5 and CD68 gene expression levels in our study subjects after adjustment for sex, age and BMI. Accordingly, MC deficiency abrogated increase in expression of pro-inflammatory M1 macrophage marker genes in mouse WAT upon high-fat diet feeding. However, MCs accumulated in obese human WAT independent of insulin resistance and systemic changes in inflammatory mediators. CONCLUSIONS: Our results suggest that MCs contribute to the local pro-inflammatory state within WAT in obesity but do not play a primary role in causing insulin resistance.


Asunto(s)
Resistencia a la Insulina/fisiología , Mastocitos/fisiología , Obesidad Mórbida/patología , Tejido Adiposo Blanco/patología , Adulto , Animales , Biomarcadores/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Citocinas/metabolismo , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Fenotipo , Delgadez/patología , Triptasas/metabolismo
7.
Nat Aging ; 4(1): 80-94, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38238601

RESUMEN

Skeletal muscle plays a central role in the regulation of systemic metabolism during lifespan. With aging, this function is perturbed, initiating multiple chronic diseases. Our knowledge of mechanisms responsible for this decline is limited. Glycerophosphocholine phosphodiesterase 1 (Gpcpd1) is a highly abundant muscle enzyme that hydrolyzes glycerophosphocholine (GPC). The physiological functions of Gpcpd1 remain largely unknown. Here we show, in mice, that the Gpcpd1-GPC metabolic pathway is perturbed in aged muscles. Further, muscle-specific, but not liver- or fat-specific, inactivation of Gpcpd1 resulted in severely impaired glucose metabolism. Western-type diets markedly worsened this condition. Mechanistically, Gpcpd1 muscle deficiency resulted in accumulation of GPC, causing an 'aged-like' transcriptomic signature and impaired insulin signaling in young Gpcpd1-deficient muscles. Finally, we report that the muscle GPC levels are markedly altered in both aged humans and patients with type 2 diabetes, displaying a high positive correlation between GPC levels and chronological age. Our findings reveal that the muscle GPCPD1-GPC metabolic pathway has an important role in the regulation of glucose homeostasis and that it is impaired during aging, which may contribute to glucose intolerance in aging.


Asunto(s)
Diabetes Mellitus Tipo 2 , Glucosa , Glicerilfosforilcolina , Fosfolipasas , Anciano , Animales , Humanos , Ratones , Envejecimiento/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Redes y Vías Metabólicas , Músculo Esquelético/metabolismo , Fosfolipasas/metabolismo , Glicerilfosforilcolina/metabolismo
8.
EMBO Mol Med ; 15(3): e16959, 2023 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-36740985

RESUMEN

The natural compound Artemisinin is the most widely used antimalarial drug worldwide. Based on its cytotoxicity, it is also used for anticancer therapy. Artemisinin and its derivates are endoperoxides that damage proteins in eukaryotic cells; their definite mechanism of action and host cell targets, however, have remained largely elusive. Using yeast and haploid stem cell screening, we demonstrate that a single cellular pathway, namely porphyrin (heme) biosynthesis, is required for the cytotoxicity of Artemisinins. Genetic or pharmacological modulation of porphyrin production is sufficient to alter its cytotoxicity in eukaryotic cells. Using multiple model systems of human brain tumor development, such as cerebral glioblastoma organoids, and patient-derived tumor spheroids, we sensitize cancer cells to dihydroartemisinin using the clinically approved porphyrin enhancer and surgical fluorescence marker 5-aminolevulinic acid, 5-ALA. A combination treatment of Artemisinins and 5-ALA markedly and specifically killed brain tumor cells in all model systems tested, including orthotopic patient-derived xenografts in vivo. These data uncover the critical molecular pathway for Artemisinin cytotoxicity and a sensitization strategy to treat different brain tumors, including drug-resistant human glioblastomas.


Asunto(s)
Antimaláricos , Artemisininas , Neoplasias Encefálicas , Humanos , Artemisininas/farmacología , Artemisininas/uso terapéutico , Antimaláricos/farmacología , Hemo/metabolismo , Ácido Aminolevulínico , Neoplasias Encefálicas/tratamiento farmacológico
9.
Nat Metab ; 5(3): 495-515, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36941451

RESUMEN

Muscle degeneration is the most prevalent cause for frailty and dependency in inherited diseases and ageing. Elucidation of pathophysiological mechanisms, as well as effective treatments for muscle diseases, represents an important goal in improving human health. Here, we show that the lipid synthesis enzyme phosphatidylethanolamine cytidyltransferase (PCYT2/ECT) is critical to muscle health. Human deficiency in PCYT2 causes a severe disease with failure to thrive and progressive weakness. pcyt2-mutant zebrafish and muscle-specific Pcyt2-knockout mice recapitulate the participant phenotypes, with failure to thrive, progressive muscle weakness and accelerated ageing. Mechanistically, muscle Pcyt2 deficiency affects cellular bioenergetics and membrane lipid bilayer structure and stability. PCYT2 activity declines in ageing muscles of mice and humans, and adeno-associated virus-based delivery of PCYT2 ameliorates muscle weakness in Pcyt2-knockout and old mice, offering a therapy for individuals with a rare disease and muscle ageing. Thus, PCYT2 plays a fundamental and conserved role in vertebrate muscle health, linking PCYT2 and PCYT2-synthesized lipids to severe muscle dystrophy and ageing.


Asunto(s)
Insuficiencia de Crecimiento , ARN Nucleotidiltransferasas , Animales , Humanos , Ratones , Ratones Noqueados , Debilidad Muscular/genética , Músculos , ARN Nucleotidiltransferasas/química , ARN Nucleotidiltransferasas/genética , Pez Cebra
10.
Sci Adv ; 8(44): eabo7247, 2022 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-36332029

RESUMEN

The HUSH (human silencing hub) complex contains the H3K9me3 binding protein M-phase phosphoprotein 8 (MPP8) and recruits the histone methyltransferase SETDB1 as well as Microrchidia CW-type zinc finger protein 2 (MORC2). Functional and mechanistic studies of the HUSH complex have hitherto been centered around SETDB1 while the in vivo functions of MPP8 and MORC2 remain elusive. Here, we show that genetic inactivation of Mphosph8 or Morc2a in the nervous system of mice leads to increased brain size, altered brain architecture, and behavioral changes. Mechanistically, in both mouse brains and human cerebral organoids, MPP8 and MORC2 suppress the repetitive-like protocadherin gene cluster in an H3K9me3-dependent manner. Our data identify MPP8 and MORC2, previously linked to silencing of repetitive elements via the HUSH complex, as key epigenetic regulators of protocadherin expression in the nervous system and thereby brain development and neuronal individuality in mice and humans.

12.
Cell Metab ; 22(1): 77-85, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26094891

RESUMEN

Diabetes results from a reduction of pancreatic ß-cells. Stimulating replication could normalize ß-cell mass. However, adult human ß-cells are recalcitrant to proliferation. We identified osteoprotegerin, a bone-related decoy receptor, as a ß-cell mitogen. Osteoprotegerin was induced by and required for lactogen-mediated rodent ß-cell replication. Osteoprotegerin enhanced ß-cell proliferation in young, aged, and diabetic mice. This resulted in increased ß-cell mass in young mice and significantly delayed hyperglycemia in diabetic mice. Osteoprotegerin stimulated replication of adult human ß-cells, without causing dedifferentiation. Mechanistically, osteoprotegerin induced human and rodent ß-cell replication by modulating CREB and GSK3 pathways, through binding Receptor Activator of NF-κB (RANK) Ligand (RANKL), a brake in ß-cell proliferation. Denosumab, an FDA-approved osteoporosis drug, and RANKL-specific antibody induced human ß-cell proliferation in vitro, and in vivo, in humanized mice. Thus, osteoprotegerin and Denosumab prevent RANKL/RANK interaction to stimulate ß-cell replication, highlighting the potential for repurposing an osteoporosis drug to treat diabetes.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Proliferación Celular/efectos de los fármacos , Denosumab/farmacología , Células Secretoras de Insulina/efectos de los fármacos , FN-kappa B/metabolismo , Osteoprotegerina/metabolismo , Animales , Línea Celular , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Humanos , Células Secretoras de Insulina/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Ligando RANK/antagonistas & inhibidores , Ratas , Transducción de Señal/efectos de los fármacos
13.
Skelet Muscle ; 3(1): 14, 2013 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-23758845

RESUMEN

BACKGROUND: Duchenne muscular dystrophy (DMD) is one of the most frequent forms of muscular disorders. It is caused by the absence of dystrophin, a core component of the sarcolemma-associated junctional complex that links the cytoskeleton to the extracellular matrix. We showed previously that plectin 1f (P1f), one of the major muscle-expressed isoforms of the cytoskeletal linker protein plectin, accumulates at the sarcolemma of DMD patients as well as of mdx mice, a widely studied animal model for DMD.Based on plectin's dual role as structural protein and scaffolding platform for signaling molecules, we speculated that the dystrophic phenotype observed after loss of dystrophin was caused, at least to some extent, by excess plectin. Thus, we hypothesized that elimination of plectin expression in mdx skeletal muscle, while probably resulting in an overall more severe phenotype, may lead to a partial phenotype rescue. In particular, we wanted to assess whether excess sarcolemmal plectin contributes to the dysregulation of sugar metabolism in mdx myofibers. METHODS: We generated plectin/dystrophin double deficient (dKO) mice by breeding mdx with conditional striated muscle-restricted plectin knockout (cKO) mice. The phenotype of these mice was comparatively analyzed with that of mdx, cKO, and wild-type mice, focusing on structural integrity and dysregulation of glucose metabolism. RESULTS: We show that the accumulation of plectin at the sarcolemma of mdx muscle fibers hardly compensated for their loss of structural integrity. Instead, it led to an additional metabolic deficit by impairing glucose uptake. While dKO mice suffered from an overall more severe form of muscular dystrophy compared to mdx or plectin-deficient mice, sarcolemmal integrity as well as glucose uptake of their myofibers were restored to normal levels upon ablation of plectin. Furthermore, microtubule (MT) networks in intact dKO myofibers, including subsarcolemmal areas, were found to be more robust than those in mdx mice. Finally, myotubes differentiated from P1f-overexpressing myoblasts showed an impairment of glucose transporter 4 translocation and a destabilization of MT networks. CONCLUSIONS: Based on these results we propose that sarcolemma-associated plectin acts as an antagonist of MT network formation in myofibers, thereby hindering vesicle-mediated (MT-dependent) transport of glucose transporter 4. This novel role of plectin throws a bridge between extra-sarcomeric cytoarchitecture and metabolism of muscle fibers. Our study thus provides new insights into pathomechanisms of plectinopathies and muscular dystrophies in general.

14.
Nat Med ; 19(3): 358-63, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23396210

RESUMEN

Hepatic insulin resistance is a driving force in the pathogenesis of type 2 diabetes mellitus (T2DM) and is tightly coupled with excessive storage of fat and the ensuing inflammation within the liver. There is compelling evidence that activation of the transcription factor nuclear factor-κB (NF-κB) and downstream inflammatory signaling pathways systemically and in the liver are key events in the etiology of hepatic insulin resistance and ß-cell dysfunction, although the molecular mechanisms involved are incompletely understood. We here test the hypothesis that receptor activator of NF-κB ligand (RANKL), a prototypic activator of NF-κB, contributes to this process using both an epidemiological and experimental approach. In the prospective population-based Bruneck Study, a high serum concentration of soluble RANKL emerged as a significant (P<0.001) and independent risk predictor of T2DM manifestation. In close agreement, systemic or hepatic blockage of RANKL signaling in genetic and nutritional mouse models of T2DM resulted in a marked improvement of hepatic insulin sensitivity and amelioration or even normalization of plasma glucose concentrations and glucose tolerance. Overall, this study provides evidence for a role of RANKL signaling in the pathogenesis of T2DM. If so, translation to the clinic may be feasible given current pharmacological strategies to lower RANKL activity to treat osteoporosis.


Asunto(s)
Diabetes Mellitus Tipo 2/prevención & control , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Ligando RANK/metabolismo , Adulto , Anciano , Animales , Línea Celular , Activación Enzimática , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Persona de Mediana Edad , FN-kappa B/metabolismo , Estudios Prospectivos , Ligando RANK/antagonistas & inhibidores
15.
Cell ; 131(3): 476-91, 2007 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-17981116

RESUMEN

Type-2 diabetes results from the development of insulin resistance and a concomitant impairment of insulin secretion. Recent studies place altered mitochondrial oxidative phosphorylation (OxPhos) as an underlying genetic element of insulin resistance. However, the causative or compensatory nature of these OxPhos changes has yet to be proven. Here, we show that muscle- and liver-specific AIF ablation in mice initiates a pattern of OxPhos deficiency closely mimicking that of human insulin resistance, and contrary to current expectations, results in increased glucose tolerance, reduced fat mass, and increased insulin sensitivity. These results are maintained upon high-fat feeding and in both genetic mosaic and ubiquitous OxPhos-deficient mutants. Importantly, the effects of AIF on glucose metabolism are acutely inducible and reversible. These findings establish that tissue-specific as well as global OxPhos defects in mice can counteract the development of insulin resistance, diabetes, and obesity.


Asunto(s)
Factor Inductor de la Apoptosis/deficiencia , Diabetes Mellitus/prevención & control , Eliminación de Gen , Marcación de Gen , Mitocondrias/metabolismo , Obesidad/prevención & control , Fosforilación Oxidativa , Animales , Factor Inductor de la Apoptosis/genética , Respiración de la Célula/efectos de los fármacos , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Dieta/efectos adversos , Glucosa/metabolismo , Insulina/farmacología , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mosaicismo/efectos de los fármacos , Músculos/citología , Músculos/efectos de los fármacos , Músculos/metabolismo , Obesidad/genética , Obesidad/metabolismo , Especificidad de Órganos/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Fenotipo , Especificidad por Sustrato/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA