Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nat Commun ; 15(1): 258, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38225272

RESUMEN

There are epidemiological associations between obesity and type 2 diabetes, cardiovascular disease and Alzheimer's disease. The role of amyloid beta 42 (Aß42) in these diverse chronic diseases is obscure. Here we show that adipose tissue releases Aß42, which is increased from adipose tissue of male mice with obesity and is associated with higher plasma Aß42. Increasing circulating Aß42 levels in male mice without obesity has no effect on systemic glucose homeostasis but has obesity-like effects on the heart, including reduced cardiac glucose clearance and impaired cardiac function. The closely related Aß40 isoform does not have these same effects on the heart. Administration of an Aß-neutralising antibody prevents obesity-induced cardiac dysfunction and hypertrophy. Furthermore, Aß-neutralising antibody administration in established obesity prevents further deterioration of cardiac function. Multi-contrast transcriptomic analyses reveal that Aß42 impacts pathways of mitochondrial metabolism and exposure of cardiomyocytes to Aß42 inhibits mitochondrial complex I. These data reveal a role for systemic Aß42 in the development of cardiac disease in obesity and suggest that therapeutics designed for Alzheimer's disease could be effective in combating obesity-induced heart failure.


Asunto(s)
Enfermedad de Alzheimer , Diabetes Mellitus Tipo 2 , Masculino , Ratones , Animales , Péptidos beta-Amiloides , Diabetes Mellitus Tipo 2/complicaciones , Anticuerpos Neutralizantes , Obesidad/complicaciones , Glucosa , Fragmentos de Péptidos
2.
Cell Death Dis ; 14(12): 787, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-38040704

RESUMEN

Lipotoxicity, the accumulation of lipids in non-adipose tissues, alters the metabolic transcriptome and mitochondrial metabolism in skeletal muscle. The mechanisms involved remain poorly understood. Here we show that lipotoxicity increased histone deacetylase 4 (HDAC4) and histone deacetylase 5 (HDAC5), which reduced the expression of metabolic genes and oxidative metabolism in skeletal muscle, resulting in increased non-oxidative glucose metabolism. This metabolic reprogramming was also associated with impaired apoptosis and ferroptosis responses, and preserved muscle cell viability in response to lipotoxicity. Mechanistically, increased HDAC4 and 5 decreased acetylation of p53 at K120, a modification required for transcriptional activation of apoptosis. Redox drivers of ferroptosis derived from oxidative metabolism were also reduced. The relevance of this pathway was demonstrated by overexpression of loss-of-function HDAC4 and HDAC5 mutants in skeletal muscle of obese db/db mice, which enhanced oxidative metabolic capacity, increased apoptosis and ferroptosis and reduced muscle mass. This study identifies HDAC4 and HDAC5 as repressors of skeletal muscle oxidative metabolism, which is linked to inhibition of cell death pathways and preservation of muscle integrity in response to lipotoxicity.


Asunto(s)
Histona Desacetilasas , Células Musculares , Ratones , Animales , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Células Musculares/metabolismo , Músculo Esquelético/metabolismo , Procesamiento Proteico-Postraduccional , Muerte Celular
3.
Elife ; 122023 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-37494090

RESUMEN

Systems genetics has begun to tackle the complexity of insulin resistance by capitalising on computational advances to study high-diversity populations. 'Diversity Outbred in Australia (DOz)' is a population of genetically unique mice with profound metabolic heterogeneity. We leveraged this variance to explore skeletal muscle's contribution to whole-body insulin action through metabolic phenotyping and skeletal muscle proteomics of 215 DOz mice. Linear modelling identified 553 proteins that associated with whole-body insulin sensitivity (Matsuda Index) including regulators of endocytosis and muscle proteostasis. To enrich for causality, we refined this network by focusing on negatively associated, genetically regulated proteins, resulting in a 76-protein fingerprint of insulin resistance. We sought to perturb this network and restore insulin action with small molecules by integrating the Broad Institute Connectivity Map platform and in vitro assays of insulin action using the Prestwick chemical library. These complementary approaches identified the antibiotic thiostrepton as an insulin resistance reversal agent. Subsequent validation in ex vivo insulin-resistant mouse muscle and palmitate-induced insulin-resistant myotubes demonstrated potent insulin action restoration, potentially via upregulation of glycolysis. This work demonstrates the value of a drug-centric framework to validate systems-level analysis by identifying potential therapeutics for insulin resistance.


Asunto(s)
Resistencia a la Insulina , Ratones , Animales , Resistencia a la Insulina/fisiología , Fibras Musculares Esqueléticas/metabolismo , Insulina/metabolismo , Músculo Esquelético/metabolismo , Proteínas/metabolismo , Variación Genética
4.
Curr Atheroscler Rep ; 20(9): 46, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30019290

RESUMEN

PURPOSE OF REVIEW: This review specifically summarises and reports terrestrial mammals of the gerbil subfamily, known as Israeli sand rats or Psammomys obesus (P. obesus) as a diet-controlled, unique, polygenic rodent model for research in the areas of obesity, type 2 diabetes, and cardiovascular diseases. The animal model closely mimics phenotypic and pathophysiological resemblance with human populations. RECENT FINDINGS: The physiological status and biochemical composition in P. obesus can be manipulated effectively by controlling its nutritional intake, making it a natural model for cardiovascular and diabetic research. Humans exhibit remarkable disparity in physiology and pathology, which are inter-dependent factors. However, variations in these factors in most animal models currently being used for cardiovascular/diabetes research are insignificant. Consequently, it is a necessity to identify and develop animal models exhibiting physiological variations mimicking human pathological conditions. We have compiled research developments conducted with this rodent model manifesting pathophysiology, closely mimicking that in human beings, thereby enabling better translation of novel therapeutic and diagnostic discoveries.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Modelos Animales de Enfermedad , Obesidad , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Cardiovasculares/terapia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Diabetes Mellitus Tipo 2/terapia , Gerbillinae , Obesidad/metabolismo , Obesidad/fisiopatología , Obesidad/terapia
5.
FASEB J ; 31(6): 2592-2602, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28258188

RESUMEN

Reciprocal regulation of hepatic glycolysis and gluconeogenesis contributes to systemic metabolic homeostasis. Recent evidence from lower order organisms has found that reversible post-translational modification of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), particularly acetylation, contributes to the reciprocal regulation of glycolysis/gluconeogenesis. However, whether this occurs in mammalian hepatocytes in vitro or in vivo is unknown. Several proteomics studies have identified 4 lysine residues in critical regions of mammalian GAPDH that are altered by multiple post-translational modifications. In FAO hepatoma cells, mutation of all 4 lysine residues (4K-R GAPDH) to mimic their unmodified state reduced GAPDH glycolytic activity and glycolytic flux and increased gluconeogenic GAPDH activity and glucose production. Hepatic expression of 4K-R GAPDH in mice increased GAPDH gluconeogenic activity and the contribution of gluconeogenesis to endogenous glucose production in the unfed state. Consistent with the increased reliance on the energy-consuming gluconeogenic pathway, plasma free fatty acids and ketones were elevated in mice expressing 4K-R GAPDH, suggesting enhanced lipolysis and hepatic fatty acid oxidation. In normal mice, food withholding and refeeding, as well as hormonal regulators of reciprocal glycolysis/gluconeogenesis, such as insulin, glucagon, and norepinephrine, had no effect on global GAPDH acetylation. However, GAPDH acetylation was reduced in obese and type 2 diabetic db/db mice. These findings show that post-translational modification of GAPDH lysine residues regulates hepatic and systemic metabolism, revealing an unappreciated role for hepatic GAPDH in substrate selection and utilization.-Bond, S. T., Howlett, K. F., Kowalski, G. M., Mason, S., Connor, T., Cooper, A., Streltsov, V., Bruce, C. R., Walder, K. R., McGee, S. L. Lysine post-translational modification of glyceraldehyde-3-phosphate dehydrogenase regulates hepatic and systemic metabolism.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Hígado/metabolismo , Lisina , Procesamiento Proteico-Postraduccional/fisiología , Secuencia de Aminoácidos , Animales , Clonación Molecular , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/genética , Ratones , Ratas
6.
Artículo en Inglés | MEDLINE | ID: mdl-28320830

RESUMEN

An acute bout of exercise is sufficient to induce changes in skeletal muscle gene expression that are ultimately responsible for the adaptive responses to exercise. Although much research has described the intracellular signaling responses to exercise that are linked to transcriptional regulation, the epigenetic mechanisms involved are only just emerging. This review will provide an overview of epigenetic mechanisms and what is known in the context of exercise. Additionally, we will explore potential interactions between metabolism during exercise and epigenetic regulation, which serves as a framework for potential areas for future research. Finally, we will consider emerging opportunities to pharmacologically manipulate epigenetic regulators and mechanisms to induce aspects of the skeletal muscle exercise adaptive response for therapeutic intervention in various disease states.


Asunto(s)
Epigénesis Genética , Ejercicio Físico/fisiología , Músculo Esquelético/fisiología , Metilación de ADN , Regulación de la Expresión Génica , Código de Histonas , Humanos , Transducción de Señal
7.
Diabetes Obes Metab ; 19(7): 936-943, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28155245

RESUMEN

AIM: To determine the effect of Scriptaid, a compound that can replicate aspects of the exercise adaptive response through disruption of the class IIa histone deacetylase (HDAC) corepressor complex, on muscle insulin action in obesity. MATERIALS AND METHODS: Diet-induced obese mice were administered Scriptaid (1 mg/kg) via daily intraperitoneal injection for 4 weeks. Whole-body and skeletal muscle metabolic phenotyping of mice was performed, in addition to echocardiography, to assess cardiac morphology and function. RESULTS: Scriptaid treatment had no effect on body weight or composition, but did increase energy expenditure, supported by increased lipid oxidation, while food intake was also increased. Scriptaid enhanced the expression of oxidative genes and proteins, increased fatty acid oxidation and reduced triglycerides and diacylglycerides in skeletal muscle. Furthermore, ex vivo insulin-stimulated glucose uptake by skeletal muscle was enhanced. Surprisingly, heart weight was reduced in Scriptaid-treated mice and was associated with enhanced expression of genes involved in oxidative metabolism in the heart. Scriptaid also improved indices of both diastolic and systolic cardiac function. CONCLUSION: These data show that pharmacological targeting of the class IIa HDAC corepressor complex with Scriptaid could be used to enhance muscle insulin action and cardiac function in obesity.


Asunto(s)
Cardiotónicos/uso terapéutico , Metabolismo Energético/efectos de los fármacos , Corazón/efectos de los fármacos , Inhibidores de Histona Desacetilasas/uso terapéutico , Hidroxilaminas/uso terapéutico , Músculo Esquelético/efectos de los fármacos , Obesidad/tratamiento farmacológico , Quinolinas/uso terapéutico , Animales , Fármacos Antiobesidad/efectos adversos , Fármacos Antiobesidad/uso terapéutico , Cardiotónicos/efectos adversos , Dieta Alta en Grasa/efectos adversos , Ecocardiografía , Ecocardiografía Doppler , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Corazón/diagnóstico por imagen , Corazón/fisiopatología , Histona Desacetilasa 2/antagonistas & inhibidores , Histona Desacetilasa 2/metabolismo , Inhibidores de Histona Desacetilasas/efectos adversos , Hidroxilaminas/efectos adversos , Resistencia a la Insulina , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Miocardio/patología , Obesidad/etiología , Obesidad/patología , Obesidad/fisiopatología , Tamaño de los Órganos , Quinolinas/efectos adversos
8.
Cell Rep ; 16(11): 2802-2810, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27626651

RESUMEN

Drugs that recapitulate aspects of the exercise adaptive response have the potential to provide better treatment for diseases associated with physical inactivity. We previously observed reduced skeletal muscle class IIa HDAC (histone deacetylase) transcriptional repressive activity during exercise. Here, we find that exercise-like adaptations are induced by skeletal muscle expression of class IIa HDAC mutants that cannot form a corepressor complex. Adaptations include increased metabolic gene expression, mitochondrial capacity, and lipid oxidation. An existing HDAC inhibitor, Scriptaid, had similar phenotypic effects through disruption of the class IIa HDAC corepressor complex. Acute Scriptaid administration to mice increased the expression of metabolic genes, which required an intact class IIa HDAC corepressor complex. Chronic Scriptaid administration increased exercise capacity, whole-body energy expenditure and lipid oxidation, and reduced fasting blood lipids and glucose. Therefore, compounds that disrupt class IIa HDAC function could be used to enhance metabolic health in chronic diseases driven by physical inactivity.


Asunto(s)
Proteínas Co-Represoras/metabolismo , Metabolismo Energético , Histona Desacetilasas/metabolismo , Metabolismo de los Lípidos , Animales , Dominio Catalítico , Línea Celular , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hidroxilaminas/administración & dosificación , Hidroxilaminas/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Ratones , Mutación/genética , Oxidación-Reducción , Condicionamiento Físico Animal , Unión Proteica/efectos de los fármacos , Quinolinas/administración & dosificación , Quinolinas/farmacología , Transcripción Genética/efectos de los fármacos
9.
Proc Natl Acad Sci U S A ; 111(39): 14153-8, 2014 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-25225394

RESUMEN

Pathogens have been hypothesized to play a major role in host diversity and speciation. Susceptibility of hybrid hosts to pathogens is thought to be a common phenomenon that could promote host population divergence and subsequently speciation. However, few studies have tested for pathogen infection across animal hybrid zones while testing for codivergence of the pathogens in the hybridizing host complex. Over 8 y, we studied natural infection by a rapidly evolving single-strand DNA virus, beak and feather diseases virus (BFDV), which infects parrots, exploiting a host-ring species complex (Platycercus elegans) in Australia. We found that host subspecies and their hybrids varied strikingly in both BFDV prevalence and load: both hybrid and phenotypically intermediate subspecies had lower prevalence and load compared with parental subspecies, while controlling for host age, sex, longitude and latitude, as well as temporal effects. We sequenced viral isolates throughout the range, which revealed patterns of genomic variation analogous to Mayr's ring-species hypothesis, to our knowledge for the first time in any host-pathogen system. Viral phylogeny, geographic location, intraspecific host density, and parrot community diversity and composition did not explain the differences in BFDV prevalence or load between subpopulations. Overall, our analyses suggest that functional host responses to infection, or force of infection, differ between subspecies and hybrids. Our findings highlight the role of host hybridization and clines in altering host-pathogen interactions, dynamics that can have important implications for models of speciation with gene flow, and offer insights into how pathogens may adapt to diverging host populations.


Asunto(s)
Enfermedades de las Aves/virología , Infecciones por Circoviridae/veterinaria , Circovirus/clasificación , Circovirus/genética , Loros/clasificación , Loros/virología , Animales , Australia , Secuencia de Bases , Infecciones por Circoviridae/virología , Circovirus/patogenicidad , ADN Viral/genética , Evolución Molecular , Femenino , Flujo Génico , Especiación Genética , Genoma Viral , Especificidad del Huésped , Interacciones Huésped-Patógeno , Hibridación Genética , Masculino , Modelos Genéticos , Datos de Secuencia Molecular , Filogenia , Carga Viral , Virulencia
10.
Cancer Chemother Pharmacol ; 71(4): 1095-100, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23420437

RESUMEN

PURPOSE: The role of v-ATPases in cancer biology is being increasingly recognized. Yeast studies indicate that the tyrosine kinase inhibitor imatinib may interact with the v-ATPase genes and alter the course of cancer progression. Data from humans in this regard are lacking. METHODS: We constructed 55 lymphoblastoid cell lines from pedigreed, cancer-free human subjects and treated them with IC20 concentration of imatinib mesylate. Using these cell lines, we (i) estimated the heritability and differential expression of 19 genes encoding several subunits of the v-ATPase protein in response to imatinib treatment; (ii) estimated the genetic similarity among these genes; and (iii) conducted a high-density scan to find cis-regulating genetic variation associated with differential expression of these genes. RESULTS: We found that the imatinib response of the genes encoding v-ATPase subunits is significantly heritable and can be clustered to identify novel drug targets in imatinib therapy. Further, five of these genes were significantly cis-regulated and together represented nearly half-log fold change in response to imatinib (p = 0.0107) that was homogenous (p = 0.2598). CONCLUSIONS: Our results proffer support to the growing view that personalized regimens using proton pump inhibitors or v-ATPase inhibitors may improve outcomes of imatinib therapy in various cancers.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Piperazinas/farmacología , Pirimidinas/farmacología , ATPasas de Translocación de Protón Vacuolares/genética , Línea Celular , Humanos , Mesilato de Imatinib , Linfocitos/efectos de los fármacos , Linfocitos/enzimología , ATPasas de Translocación de Protón Vacuolares/fisiología
11.
BMC Med Genomics ; 5: 37, 2012 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-22917222

RESUMEN

BACKGROUND: Imatinib mesylate is currently the drug of choice to treat chronic myeloid leukemia. However, patient resistance and cytotoxicity make secondary lines of treatment, such as omacetaxine mepesuccinate, a necessity. Given that drug cytotoxicity represents a major problem during treatment, it is essential to understand the biological pathways affected to better predict poor drug response and prioritize a treatment regime. METHODS: We conducted cell viability and gene expression assays to determine heritability and gene expression changes associated with imatinib and omacetaxine treatment of 55 non-cancerous lymphoblastoid cell lines, derived from 17 pedigrees. In total, 48,803 transcripts derived from Illumina Human WG-6 BeadChips were analyzed for each sample using SOLAR, whilst correcting for kinship structure. RESULTS: Cytotoxicity within cell lines was highly heritable following imatinib treatment (h2 = 0.60-0.73), but not omacetaxine treatment. Cell lines treated with an IC20 dose of imatinib or omacetaxine showed differential gene expression for 956 (1.96%) and 3,892 transcripts (7.97%), respectively; 395 of these (0.8%) were significantly influenced by both imatinib and omacetaxine treatment. k-means clustering and DAVID functional annotation showed expression changes in genes related to kinase binding and vacuole-related functions following imatinib treatment, whilst expression changes in genes related to cell division and apoptosis were evident following treatment with omacetaxine. The enrichment scores for these ontologies were very high (mostly >10). CONCLUSIONS: Induction of gene expression changes related to different pathways following imatinib and omacetaxine treatment suggests that the cytotoxicity of such drugs may be differentially tolerated by individuals based on their genetic background.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Harringtoninas/toxicidad , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Piperazinas/toxicidad , Pirimidinas/toxicidad , Benzamidas , Línea Celular , Análisis por Conglomerados , Relación Dosis-Respuesta a Droga , Homoharringtonina , Humanos , Mesilato de Imatinib , Patrón de Herencia/genética , Anotación de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Reproducibilidad de los Resultados
12.
Diabetes ; 61(8): 2146-54, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22586591

RESUMEN

We previously used Gene Expression Signature technology to identify methazolamide (MTZ) and related compounds with insulin sensitizing activity in vitro. The effects of these compounds were investigated in diabetic db/db mice, insulin-resistant diet-induced obese (DIO) mice, and rats with streptozotocin (STZ)-induced diabetes. MTZ reduced fasting blood glucose and HbA(1c) levels in db/db mice, improved glucose tolerance in DIO mice, and enhanced the glucose-lowering effects of exogenous insulin administration in rats with STZ-induced diabetes. Hyperinsulinemic-euglycemic clamps in DIO mice revealed that MTZ increased glucose infusion rate and suppressed endogenous glucose production. Whole-body or cellular oxygen consumption rate was not altered, suggesting MTZ may inhibit glucose production by different mechanism(s) to metformin. In support of this, MTZ enhanced the glucose-lowering effects of metformin in db/db mice. MTZ is known to be a carbonic anhydrase inhibitor (CAI); however, CAIs acetazolamide, ethoxyzolamide, dichlorphenamide, chlorthalidone, and furosemide were not effective in vivo. Our results demonstrate that MTZ acts as an insulin sensitizer that suppresses hepatic glucose production in vivo. The antidiabetic effect of MTZ does not appear to be a function of its known activity as a CAI. The additive glucose-lowering effect of MTZ together with metformin highlights the potential utility for the management of type 2 diabetes.


Asunto(s)
Glucemia/efectos de los fármacos , Hipoglucemiantes/uso terapéutico , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Metazolamida/uso terapéutico , Animales , Glucemia/metabolismo , Inhibidores de Anhidrasa Carbónica/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Técnica de Clampeo de la Glucosa , Glucosa-6-Fosfatasa/efectos de los fármacos , Glucólisis/efectos de los fármacos , Homeostasis/efectos de los fármacos , Insulina/uso terapéutico , Masculino , Metformina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Consumo de Oxígeno/efectos de los fármacos , Fosfoenolpiruvato Carboxiquinasa (ATP)/efectos de los fármacos , Ácido Pirúvico/metabolismo , Ratas , Ratas Sprague-Dawley
13.
Physiol Genomics ; 43(3): 110-20, 2011 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-21081660

RESUMEN

Insulin resistance is a heterogeneous disorder caused by a range of genetic and environmental factors, and we hypothesize that its etiology varies considerably between individuals. This heterogeneity provides significant challenges to the development of effective therapeutic regimes for long-term management of type 2 diabetes. We describe a novel strategy, using large-scale gene expression profiling, to develop a gene expression signature (GES) that reflects the overall state of insulin resistance in cells and patients. The GES was developed from 3T3-L1 adipocytes that were made "insulin resistant" by treatment with tumor necrosis factor-α (TNF-α) and then reversed with aspirin and troglitazone ("resensitized"). The GES consisted of five genes whose expression levels best discriminated between the insulin-resistant and insulin-resensitized states. We then used this GES to screen a compound library for agents that affected the GES genes in 3T3-L1 adipocytes in a way that most closely resembled the changes seen when insulin resistance was successfully reversed with aspirin and troglitazone. This screen identified both known and new insulin-sensitizing compounds including nonsteroidal anti-inflammatory agents, ß-adrenergic antagonists, ß-lactams, and sodium channel blockers. We tested the biological relevance of this GES in participants in the San Antonio Family Heart Study (n = 1,240) and showed that patients with the lowest GES scores were more insulin resistant (according to HOMA_IR and fasting plasma insulin levels; P < 0.001). These findings show that GES technology can be used for both the discovery of insulin-sensitizing compounds and the characterization of patients into subtypes of insulin resistance according to GES scores, opening the possibility of developing a personalized medicine approach to type 2 diabetes.


Asunto(s)
Perfilación de la Expresión Génica , Resistencia a la Insulina/genética , Células 3T3-L1 , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Insulina/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Transporte de Proteínas/efectos de los fármacos , Reproducibilidad de los Resultados , Factor de Necrosis Tumoral alfa/farmacología , Adulto Joven
14.
Metabolism ; 59(7): 1057-64, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20045148

RESUMEN

The human Abelson helper integration site-1 (AHI1) gene is associated with both neurologic and hematologic disorders; however, it is also located in a chromosomal region linked to metabolic syndrome phenotypes and was identified as a type 2 diabetes mellitus susceptibility gene from a genomewide association study. To further define a possible role in type 2 diabetes mellitus development, AHI1 messenger RNA expression levels were investigated in a range of tissues and found to be highly expressed in skeletal muscle as well as displaying elevated levels in brain regions and gonad tissues. Further analysis in a rodent polygenic animal model of obesity and type 2 diabetes mellitus identified increased Ahi-1 messenger RNA levels in red gastrocnemius muscle from fasted impaired glucose-tolerant and diabetic rodents compared with healthy animals (P < .002). Moreover, elevated gene expression levels were confirmed in skeletal muscle from fasted obese and type 2 diabetes mellitus human subjects (P < .02). RNAi-mediated suppression of Ahi-1 resulted in increased glucose transport in rat L6 myotubes in both the basal and insulin-stimulated states (P < .01). Finally, single nucleotide polymorphism association studies identified 2 novel AHI1 genetic variants linked with fasting blood glucose levels in Mexican American subjects (P < .037). These findings indicate a novel role for AHI1 in skeletal muscle and identify additional genetic links with metabolic syndrome phenotypes suggesting an involvement of AHI1 in the maintenance of glucose homeostasis and type 2 diabetes mellitus progression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Síndrome Metabólico/metabolismo , Músculo Esquelético/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras del Transporte Vesicular , Animales , Glucemia/metabolismo , Western Blotting , Peso Corporal/fisiología , Células Cultivadas , Estudios de Cohortes , Desoxiglucosa/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Genotipo , Glucosa/metabolismo , Humanos , Insulina/sangre , Resistencia a la Insulina/genética , Síndrome Metabólico/genética , Americanos Mexicanos , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Obesidad/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
15.
Hum Genet ; 127(2): 183-90, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19862556

RESUMEN

Given their involvement in processes necessary for life, mitochondrial damage and subsequent dysfunction can lead to a wide range of human diseases. Previous studies of both animal models and humans have suggested that presenilins-associated rhomboid-like protein (PARL) is a key regulator of mitochondrial integrity and function, and plays a role in cellular apoptosis. As a surrogate measure of mitochondrial integrity, we previously measured mitochondrial content in a Caucasian population consisting of large extended pedigrees, with results highlighting a substantial genetic component to this trait. To assess the influence of variation in the PARL gene on mitochondrial content, we re-sequenced 6.5 kb of the gene, identifying 16 SNPs and genotyped these in 1,086 Caucasian individuals, distributed across 170 families. Statistical genetic analysis revealed that one promoter variant, T-191C, exhibited significant effects (after correction for multiple testing) on mitochondrial content levels. Comparison of the transcription factor binding characteristics of the T-191C promoter SNP by EMSA indicates preferential binding of nuclear factors to the T allele, suggesting functional variation in PARL expression. These results suggest that genetic variation within PARL influences mitochondrial abundance and integrity.


Asunto(s)
ADN Mitocondrial/genética , Metaloproteasas/genética , Proteínas Mitocondriales/genética , Polimorfismo de Nucleótido Simple , ADN Mitocondrial/química , Salud de la Familia , Femenino , Frecuencia de los Genes , Variación Genética , Genotipo , Humanos , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas/genética , Análisis de Secuencia de ADN , Población Blanca/genética
16.
J Physiol ; 587(Pt 7): 1619-34, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19204049

RESUMEN

Skeletal muscle tissue undergoes adaptive changes in response to stress and the genes that control these processes are incompletely characterised. NDRG2 (N-myc downstream-regulated gene 2), a stress- and growth-related gene, was investigated in skeletal muscle growth and adaption. While NDRG2 expression levels were found to be up-regulated in both differentiated human and mouse myotubes compared with undifferentiated myoblasts, the suppression of NDRG2 in C2C12 myoblasts resulted in slowed myoblast proliferation. The increased expression levels of the cell cycle inhibitors, p21 Waf1/Cip1 and p27 Kip1, and of various muscle differentiation markers in NDRG2-deficient myoblasts indicate that a lack of NDRG2 promoted cell cycle exiting and the onset of myogenesis. Furthermore, the analysis of NDRG2 regulation in C2C12 myotubes treated with catabolic and anabolic agents and in skeletal muscle from human subjects following resistance exercise training revealed NDRG2 gene expression to be down-regulated during hypertrophic conditions, and conversely, up-regulated during muscle atrophy. Together, these data demonstrate that NDRG2 expression is highly responsive to different stress conditions in skeletal muscle and suggest that the level of NDRG2 expression may be critical to myoblast growth and differentiation.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Desarrollo de Músculos , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Mioblastos Esqueléticos/metabolismo , Proteínas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Factores de Edad , Anciano , Animales , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Supervivencia Celular , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Humanos , Hipertrofia , Masculino , Ratones , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/patología , Mioblastos Esqueléticos/patología , Fenotipo , Proteínas/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Entrenamiento de Fuerza , Proteínas Ligasas SKP Cullina F-box/metabolismo , Factores de Tiempo , Transfección , Proteínas de Motivos Tripartitos , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/metabolismo , Adulto Joven
17.
Hum Biol ; 78(2): 147-59, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17036923

RESUMEN

The BEACON gene was initially identified using the differential display polymerase chain reaction on hypothalamic mRNA samples collected from lean and obese Psammomys obesus, a polygenic animal model of obesity. Hypothalamic BEACON gene expression was positively correlated with percentage of body fat, and intracerebroventricular infusion of the Beacon protein resulted in a dose-dependent increase in food intake and body weight. The human homolog of BEACON, UBL5, is located on chromosome 19p in a region previously linked to quantitative traits related to obesity. Our previous studies showed a statistically significant association between UBL5 sequence variation and several obesity- and diabetes-related quantitative physiological measures in Asian Indian and Micronesian cohorts. Here we undertake a replication study in a Mexican American cohort where the original linkage signal was first detected. We exhaustively resequenced the complete gene plus the putative promoter region for genetic variation in 55 individuals and identified five single nucleotide polymorphisms (SNPs), one of which was novel. These SNPs were genotyped in a Mexican American cohort of 900 individuals from 40 families. Using a quantitative trait linkage disequilibrium test, we found significant associations between UBL5 genetic variants and waist-to-hip ratio (p = 0.027), and the circulating concentrations of insulin (p = 0.018) and total cholesterol (p = 0.023) in fasted individuals. These data are consistent with our earlier published studies and further support a functional role for the UBL5 gene in influencing physiological traits that underpin the development of metabolic syndrome.


Asunto(s)
Proteínas del Ojo/genética , Variación Genética/genética , Genética de Población/métodos , Síndrome Metabólico/genética , Americanos Mexicanos , Ubiquitinas/genética , Adulto , Diabetes Mellitus/genética , Femenino , Humanos , Masculino , Obesidad/genética , Polimorfismo de Nucleótido Simple/genética , Texas
18.
Nat Genet ; 37(11): 1234-41, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16227999

RESUMEN

Chronic inflammation has a pathological role in many common diseases and is influenced by both genetic and environmental factors. Here we assess the role of genetic variation in selenoprotein S (SEPS1, also called SELS or SELENOS), a gene involved in stress response in the endoplasmic reticulum and inflammation control. After resequencing SEPS1, we genotyped 13 SNPs in 522 individuals from 92 families. As inflammation biomarkers, we measured plasma levels of IL-6, IL-1beta and TNF-alpha. Bayesian quantitative trait nucleotide analysis identified associations between SEPS1 polymorphisms and all three proinflammatory cytokines. One promoter variant, -105G --> A, showed strong evidence for an association with each cytokine (multivariate P = 0.0000002). Functional analysis of this polymorphism showed that the A variant significantly impaired SEPS1 expression after exposure to endoplasmic reticulum stress agents (P = 0.00006). Furthermore, suppression of SEPS1 by short interfering RNA in macrophage cells increased the release of IL-6 and TNF-alpha. To investigate further the significance of the observed associations, we genotyped -105G --> A in 419 Mexican American individuals from 23 families for replication. This analysis confirmed a significant association with both TNF-alpha (P = 0.0049) and IL-1beta (P = 0.0101). These results provide a direct mechanistic link between SEPS1 and the production of inflammatory cytokines and suggest that SEPS1 has a role in mediating inflammation.


Asunto(s)
Variación Genética , Inflamación/genética , Proteínas de la Membrana/genética , Polimorfismo de Nucleótido Simple/genética , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Genotipo , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-1/sangre , Interleucina-6/sangre , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Regiones Promotoras Genéticas/genética , ARN Interferente Pequeño/farmacología , Selenoproteínas , Factor de Necrosis Tumoral alfa/metabolismo
19.
Diabetes ; 53(9): 2467-72, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15331561

RESUMEN

The BEACON gene (also known as UBL5) was identified as differentially expressed between lean and obese Psammomys obesus, a polygenic animal model of obesity, type 2 diabetes, and dyslipidemia. The human homologue of BEACON is located on chromosome 19p, a region likely to contain genes affecting metabolic syndrome-related quantitative traits as established by linkage studies. To assess whether the human BEACON gene may be involved in influencing these traits, we exhaustively analyzed the complete gene for genetic variation in 40 unrelated individuals and identified four variants (three novel). The two more common variants were tested for association with a number of quantitative metabolic syndrome-related traits in two large cohorts of unrelated individuals. Significant associations were found between these variants and fat mass (P = 0.026), percentage of fat (P = 0.001), and waist-to-hip ratio (P = 0.031). The same variants were also associated with total cholesterol (P = 0.024), LDL cholesterol (P = 0.019), triglycerides (P = 0.006), and postglucose load insulin levels (P = 0.018). Multivariate analysis of these correlated phenotypes also yielded a highly significant association (P = 0.0004), suggesting that BEACON may influence phenotypic variation in metabolic syndrome-related traits.


Asunto(s)
Proteínas del Ojo/genética , Síndrome Metabólico/etnología , Síndrome Metabólico/genética , Ubiquitinas/genética , Adulto , Diabetes Mellitus/etnología , Diabetes Mellitus/genética , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Hiperlipidemias/etnología , Hiperlipidemias/genética , Masculino , Mauricio/epidemiología , Persona de Mediana Edad , Obesidad , Fenotipo , Prevalencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...