Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Metab ; 24: 1-17, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31003944

RESUMEN

OBJECTIVE: Aging is accompanied by loss of brown adipocytes and a decline in their thermogenic potential, which may exacerbate the development of adiposity and other metabolic disorders. Presently, only limited evidence exists describing the molecular alterations leading to impaired brown adipogenesis with aging and the contribution of these processes to changes of systemic energy metabolism. METHODS: Samples of young and aged murine brown and white adipose tissue were used to compare age-related changes of brown adipogenic gene expression and thermogenesis-related lipid mobilization. To identify potential markers of brown adipose tissue aging, non-targeted proteomic and metabolomic as well as targeted lipid analyses were conducted on young and aged tissue samples. Subsequently, the effects of several candidate lipid classes on brown adipocyte function were examined. RESULTS: Corroborating previous reports of reduced expression of uncoupling protein-1, we observe impaired signaling required for lipid mobilization in aged brown fat after adrenergic stimulation. Omics analyses additionally confirm the age-related impairment of lipid homeostasis and reveal the accumulation of specific lipid classes, including certain sphingolipids, ceramides, and dolichols in aged brown fat. While ceramides as well as enzymes of dolichol metabolism inhibit brown adipogenesis, inhibition of sphingosine 1-phosphate receptor 2 induces brown adipocyte differentiation. CONCLUSIONS: Our functional analyses show that changes in specific lipid species, as observed during aging, may contribute to reduced thermogenic potential. They thus uncover potential biomarkers of aging as well as molecular mechanisms that could contribute to the degradation of brown adipocytes, thereby providing potential treatment strategies of age-related metabolic conditions.


Asunto(s)
Adipocitos Marrones/metabolismo , Envejecimiento/metabolismo , Metabolismo de los Lípidos , Adipocitos Marrones/citología , Animales , Biomarcadores/metabolismo , Células Cultivadas , Ceramidas/metabolismo , Dolicoles/metabolismo , Masculino , Metaboloma , Ratones , Ratones Endogámicos C57BL , Proteoma/genética , Proteoma/metabolismo , Esfingolípidos/metabolismo
2.
Aging Cell ; 16(4): 761-772, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28544360

RESUMEN

Impaired insulin/IGF1 signalling has been shown to extend lifespan in model organisms ranging from yeast to mammals. Here we sought to determine the effect of targeted disruption of the insulin receptor (IR) in non-neuronal tissues of adult mice on the lifespan. We induced hemizygous (PerIRKO+/- ) or homozygous (PerIRKO-/- ) disruption of the IR in peripheral tissue of 15-weeks-old mice using a tamoxifen-inducible Cre transgenic mouse with only peripheral tissue expression, and subsequently monitored glucose metabolism, insulin signalling and spontaneous death rates over 4 years. Complete peripheral IR disruption resulted in a diabetic phenotype with increased blood glucose and plasma insulin levels in young mice. Although blood glucose levels returned to normal, and fat mass was reduced in aged PerIRKO-/- mice, their lifespan was reduced. By contrast, heterozygous disruption had no effect on lifespan. This was despite young male PerIRKO+/- mice showing reduced fat mass and mild increase in hepatic insulin sensitivity. In conflict with findings in metazoans like Caenorhabditis elegans and Drosophila melanogaster, our results suggest that heterozygous impairment of the insulin signalling limited to peripheral tissues of adult mice fails to extend lifespan despite increased systemic insulin sensitivity, while homozygous impairment shortens lifespan.


Asunto(s)
Tejido Adiposo/metabolismo , Insulina/metabolismo , Longevidad/genética , Receptor de Insulina/genética , Transducción de Señal , Animales , Glucemia/metabolismo , Expresión Génica , Heterocigoto , Homocigoto , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Noqueados , Mutación , Receptor de Insulina/deficiencia
3.
Endocrinology ; 157(7): 2735-49, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27183316

RESUMEN

p53 is a well-known tumor suppressor that plays multiple biological roles, including the capacity to modulate metabolism at different levels. However, its metabolic role in brown adipose tissue (BAT) remains largely unknown. Herein we sought to investigate the physiological role of endogenous p53 in BAT and its implication on BAT thermogenic activity and energy balance. To this end, we generated and characterized global p53-null mice and mice lacking p53 specifically in BAT. Additionally we performed gain-and-loss-of-function experiments in the BAT of adult mice using virogenetic and pharmacological approaches. BAT was collected and analyzed by immunohistochemistry, thermography, real-time PCR, and Western blot. p53-deficient mice were resistant to diet-induced obesity due to increased energy expenditure and BAT activity. However, the deletion of p53 in BAT using a Myf5-Cre driven p53 knockout did not show any changes in body weight or the expression of thermogenic markers. The acute inhibition of p53 in the BAT of adult mice slightly increased body weight and inhibited BAT thermogenesis, whereas its overexpression in the BAT of diet-induced obese mice reduced body weight and increased thermogenesis. On the other hand, pharmacological activation of p53 improves body weight gain due to increased BAT thermogenesis by sympathetic nervous system in obese adult wild-type mice but not in p53(-/-) animals. These results reveal that p53 regulates BAT metabolism by coordinating body weight and thermogenesis, but these metabolic actions are tissue specific and also dependent on the developmental stage.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Obesidad/genética , Termogénesis/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Tejido Adiposo Pardo/metabolismo , Animales , Composición Corporal/efectos de los fármacos , Composición Corporal/genética , Peso Corporal/genética , Línea Celular , Doxorrubicina/farmacología , Masculino , Ratones , Ratones Noqueados , Obesidad/metabolismo , Ratas , Somatotrofos/citología , Somatotrofos/efectos de los fármacos , Somatotrofos/metabolismo , Termogénesis/genética , Proteína p53 Supresora de Tumor/agonistas , Proteína p53 Supresora de Tumor/metabolismo
4.
Nat Commun ; 6: 10043, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26620638

RESUMEN

Ageing has been defined as a global decline in physiological function depending on both environmental and genetic factors. Here we identify gene transcripts that are similarly regulated during physiological ageing in nematodes, zebrafish and mice. We observe the strongest extension of lifespan when impairing expression of the branched-chain amino acid transferase-1 (bcat-1) gene in C. elegans, which leads to excessive levels of branched-chain amino acids (BCAAs). We further show that BCAAs reduce a LET-363/mTOR-dependent neuro-endocrine signal, which we identify as DAF-7/TGFß, and that impacts lifespan depending on its related receptors, DAF-1 and DAF-4, as well as ultimately on DAF-16/FoxO and HSF-1 in a cell-non-autonomous manner. The transcription factor HLH-15 controls and epistatically synergizes with BCAT-1 to modulate physiological ageing. Lastly and consistent with previous findings in rodents, nutritional supplementation of BCAAs extends nematodal lifespan. Taken together, BCAAs act as periphery-derived metabokines that induce a central neuro-endocrine response, culminating in extended healthspan.


Asunto(s)
Envejecimiento/metabolismo , Aminoácidos de Cadena Ramificada/metabolismo , Caenorhabditis elegans/metabolismo , Envejecimiento/genética , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Femenino , Longevidad , Masculino , Ratones/genética , Ratones/crecimiento & desarrollo , Ratones/metabolismo , Ratones Endogámicos C57BL , Transaminasas/genética , Transaminasas/metabolismo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
5.
Nat Commun ; 5: 3563, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24714520

RESUMEN

D-Glucosamine (GlcN) is a freely available and commonly used dietary supplement potentially promoting cartilage health in humans, which also acts as an inhibitor of glycolysis. Here we show that GlcN, independent of the hexosamine pathway, extends Caenorhabditis elegans life span by impairing glucose metabolism that activates AMP-activated protein kinase (AMPK/AAK-2) and increases mitochondrial biogenesis. Consistent with the concept of mitohormesis, GlcN promotes increased formation of mitochondrial reactive oxygen species (ROS) culminating in increased expression of the nematodal amino acid-transporter 1 (aat-1) gene. Ameliorating mitochondrial ROS formation or impairment of aat-1-expression abolishes GlcN-mediated life span extension in an NRF2/SKN-1-dependent fashion. Unlike other calorie restriction mimetics, such as 2-deoxyglucose, GlcN extends life span of ageing C57BL/6 mice, which show an induction of mitochondrial biogenesis, lowered blood glucose levels, enhanced expression of several murine amino-acid transporters, as well as increased amino-acid catabolism. Taken together, we provide evidence that GlcN extends life span in evolutionary distinct species by mimicking a low-carbohydrate diet.


Asunto(s)
Envejecimiento/efectos de los fármacos , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/fisiología , Glucosamina/farmacología , Longevidad/efectos de los fármacos , Animales , Femenino , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
6.
Nat Chem Biol ; 9(11): 693-700, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24077178

RESUMEN

Sirtuins, a family of histone deacetylases, have a fiercely debated role in regulating lifespan. In contrast with recent observations, here we find that overexpression of sir-2.1, the ortholog of mammalian SirT1, does extend Caenorhabditis elegans lifespan. Sirtuins mandatorily convert NAD(+) into nicotinamide (NAM). We here find that NAM and its metabolite, 1-methylnicotinamide (MNA), extend C. elegans lifespan, even in the absence of sir-2.1. We identify a previously unknown C. elegans nicotinamide-N-methyltransferase, encoded by a gene now named anmt-1, to generate MNA from NAM. Disruption and overexpression of anmt-1 have opposing effects on lifespan independent of sirtuins, with loss of anmt-1 fully inhibiting sir-2.1-mediated lifespan extension. MNA serves as a substrate for a newly identified aldehyde oxidase, GAD-3, to generate hydrogen peroxide, which acts as a mitohormetic reactive oxygen species signal to promote C. elegans longevity. Taken together, sirtuin-mediated lifespan extension depends on methylation of NAM, providing an unexpected mechanistic role for sirtuins beyond histone deacetylation.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiología , Longevidad , Niacinamida/metabolismo , Sirtuinas/metabolismo , Animales , Caenorhabditis elegans/metabolismo , Metilación , Niacinamida/química , Sirtuinas/genética
7.
Aging Cell ; 12(3): 508-17, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23534459

RESUMEN

Arsenite is one of the most toxic chemical substances known and is assumed to exert detrimental effects on viability even at lowest concentrations. By contrast and unlike higher concentrations, we here find that exposure to low-dose arsenite promotes growth of cultured mammalian cells. In the nematode C. elegans, low-dose arsenite promotes resistance against thermal and chemical stressors and extends lifespan of this metazoan, whereas higher concentrations reduce longevity. While arsenite causes a transient increase in reactive oxygen species (ROS) levels in C. elegans, co-exposure to ROS scavengers prevents the lifespan-extending capabilities of arsenite, indicating that transiently increased ROS levels act as transducers of arsenite effects on lifespan, a process known as mitohormesis. This requires two transcription factors, namely DAF-16 and SKN-1, which employ the metallothionein MTL-2 as well as the mitochondrial transporter TIN-9.1 to extend lifespan. Taken together, low-dose arsenite extends lifespan, providing evidence for nonlinear dose-response characteristics of toxin-mediated stress resistance and longevity in a multicellular organism.


Asunto(s)
Arsenitos/farmacología , Caenorhabditis elegans/efectos de los fármacos , Hormesis , Longevidad/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Teratógenos/farmacología , Células 3T3 , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Línea Celular , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción Forkhead , Células Hep G2 , Humanos , Metalotioneína/metabolismo , Ratones , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno , Superóxido Dismutasa/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética
8.
Cell Metab ; 15(4): 451-65, 2012 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-22482728

RESUMEN

Impaired insulin and IGF-1 signaling (iIIS) in C. elegans daf-2 mutants extends life span more than 2-fold. Constitutively, iIIS increases mitochondrial activity and reduces reactive oxygen species (ROS) levels. By contrast, acute impairment of daf-2 in adult C. elegans reduces glucose uptake and transiently increases ROS. Consistent with the concept of mitohormesis, this ROS signal causes an adaptive response by inducing ROS defense enzymes (SOD, catalase), culminating in ultimately reduced ROS levels despite increased mitochondrial activity. Inhibition of this ROS signal by antioxidants reduces iIIS-mediated longevity by up to 60%. Induction of the ROS signal requires AAK-2 (AMPK), while PMK-1 (p38) and SKN-1 (NRF-2) are needed for the retrograde response. IIIS upregulates mitochondrial L-proline catabolism, and impairment of the latter impairs the life span-extending capacity of iIIS while L-proline supplementation extends C. elegans life span. Taken together, iIIS promotes L-proline metabolism to generate a ROS signal for the adaptive induction of endogenous stress defense to extend life span.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/metabolismo , Insulina/metabolismo , Longevidad/fisiología , Mitocondrias/metabolismo , Prolina/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Animales , Antioxidantes/farmacología , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Perfilación de la Expresión Génica , Glucosa/metabolismo , Longevidad/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Modelos Animales , Transducción de Señal/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Transcriptoma/genética
9.
Proc Biol Sci ; 278(1724): 3490-6, 2011 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-21490011

RESUMEN

High blood glucose levels caused by excessive sugar consumption are detrimental to mammalian health and life expectancy. Despite consuming vast quantities of sugar-rich floral nectar, nectar-feeding bats are long-lived, provoking the question of how they regulate blood glucose. We investigated blood glucose levels in nectar-feeding bats (Glossophaga soricina) in experiments in which we varied the amount of dietary sugar or flight time. Blood glucose levels increased with the quantity of glucose ingested and exceeded 25 mmol l(-1) blood in resting bats, which is among the highest values ever recorded in mammals fed sugar quantities similar to their natural diet. During normal feeding, blood glucose values decreased with increasing flight time, but only fell to expected values when bats spent 75 per cent of their time airborne. Either nectar-feeding bats have evolved mechanisms to avoid negative health effects of hyperglycaemia, or high activity is key to balancing blood glucose levels during foraging. We suggest that the coevolutionary specialization of bats towards a nectar diet was supported by the high activity and elevated metabolic rates of these bats. High activity may have conferred benefits to the bats in terms of behavioural interactions and foraging success, and is simultaneously likely to have increased their efficiency as plant pollinators.


Asunto(s)
Glucemia/análisis , Glucemia/metabolismo , Quirópteros/metabolismo , Actividad Motora , Animales , Quirópteros/fisiología , Dieta , Carbohidratos de la Dieta/análisis , Carbohidratos de la Dieta/metabolismo , Metabolismo Energético , Prueba de Tolerancia a la Glucosa/veterinaria , Néctar de las Plantas/análisis , Néctar de las Plantas/metabolismo , Factores de Tiempo
10.
Aging (Albany NY) ; 2(11): 843-53, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21084725

RESUMEN

Cardiac failure is the most prevalent cause of death at higher age, and is commonly associated with impaired energy homeostasis in the heart. Mitochondrial metabolism appears critical to sustain cardiac function to counteract aging. In this study, we generated mice transgenically over-expressing the mitochondrial protein frataxin, which promotes mitochondrial energy conversion by controlling iron-sulfur-cluster biogenesis and hereby mitochondrial electron flux. Hearts of transgenic mice displayed increased mitochondrial energy metabolism and induced stress defense mechanisms, while overall oxidative stress was decreased. Following standardized exposure to doxorubicin to induce experimental cardiomyopathy, cardiac function and survival was significantly improved in the transgenic mice. The insulin/IGF-1 signaling cascade is an important pathway that regulates survival following cytotoxic stress through the downstream targets protein kinase B, Akt, and glycogen synthase kinase 3. Activation of this cascade is markedly inhibited in the hearts of wild-type mice following induction of cardiomyopathy. By contrast, transgenic overexpression of frataxin rescues impaired insulin/IGF-1 signaling and provides a mechanism to explain enhanced cardiac stress resistance in transgenic mice. Taken together, these findings suggest that increased mitochondrial metabolism elicits an adaptive response due to mildly increased oxidative stress as a consequence of increased oxidative energy conversion, previously named mitohormesis. This in turn activates protective mechanisms which counteract cardiotoxic stress and promote survival in states of experimental cardiomyopathy. Thus, induction of mitochondrial metabolism may be considered part of a generally protective mechanism to prevent cardiomyopathy and cardiac failure.


Asunto(s)
Metabolismo Energético , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/prevención & control , Mitocondrias/metabolismo , Animales , Antibióticos Antineoplásicos/efectos adversos , Modelos Animales de Enfermedad , Doxorrubicina/efectos adversos , Insuficiencia Cardíaca/inducido químicamente , Hemodinámica , Humanos , Insulina/metabolismo , Proteínas de Unión a Hierro/metabolismo , Ratones , Ratones Transgénicos , Transducción de Señal/fisiología , Frataxina
11.
Aging (Albany NY) ; 2(10): 650-8, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20876939

RESUMEN

Reduced telomere length and impaired telomerase activity have been linked to several diseases associated with senescence and aging. However, a causal link to metabolic disorders and in particular diabetes mellitus is pending. We here show that young adult mice which are deficient for the Terc subunit of telomerase exhibit impaired glucose tolerance. This is caused by impaired glucose-stimulated insulin secretion (GSIS) from pancreatic islets, while body fat content, energy expenditure and insulin sensitivity were found to be unaltered. The impaired secretion capacity for insulin is due to reduced islet size which is linked to an impaired replication capacity of insulin-producing beta-cells in Terc-deficient mice. Taken together, telomerase deficiency and hence short telomeres impair replicative capacity of pancreatic beta-cells to cause impaired insulin secretion and glucose intolerance, mechanistically defining diabetes mellitus as an aging-associated disorder.


Asunto(s)
Glucemia/metabolismo , Intolerancia a la Glucosa/genética , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Telomerasa/deficiencia , Animales , Glucemia/efectos de los fármacos , Composición Corporal/genética , Dióxido de Carbono/metabolismo , Proliferación Celular , Metabolismo Energético/genética , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Insulina/farmacología , Células Secretoras de Insulina/patología , Islotes Pancreáticos/patología , Lípidos/sangre , Ratones , Ratones Noqueados , Oxidación-Reducción , ARN/genética , Telomerasa/genética , Telómero/metabolismo
12.
Biochem J ; 432(1): 165-72, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20819074

RESUMEN

DNA-repair mechanisms enable cells to maintain their genetic information by protecting it from mutations that may cause malignant growth. Recent evidence suggests that specific DNA-repair enzymes contain ISCs (iron-sulfur clusters). The nuclearencoded protein frataxin is essential for the mitochondrial biosynthesis of ISCs. Frataxin deficiency causes a neurodegenerative disorder named Friedreich's ataxia in humans. Various types of cancer occurring at young age are associated with this disease, and hence with frataxin deficiency. Mice carrying a hepatocyte-specific disruption of the frataxin gene develop multiple liver tumours for unresolved reasons. In the present study, we show that frataxin deficiency in murine liver is associated with increased basal levels of oxidative DNA base damage. Accordingly, eukaryotic V79 fibroblasts overexpressing human frataxin show decreased basal levels of these modifications, while prokaryotic Salmonella enterica serotype Typhimurium TA104 strains transformed with human frataxin show decreased mutation rates. The repair rates of oxidative DNA base modifications in V79 cells overexpressing frataxin were significantly higher than in control cells. Lastly, cleavage activity related to the ISC-independent repair enzyme 8-oxoguanine glycosylase was found to be unaltered by frataxin overexpression. These findings indicate that frataxin modulates DNA-repair mechanisms probably due to its impact on ISC-dependent repair proteins, linking mitochondrial dysfunction to DNA repair and tumour initiation.


Asunto(s)
Daño del ADN , Reparación del ADN/genética , Ataxia de Friedreich/genética , Proteínas de Unión a Hierro/genética , Animales , Línea Celular , Células Cultivadas , ADN Glicosilasas/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Ataxia de Friedreich/metabolismo , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Proteínas de Unión a Hierro/metabolismo , Proteínas Hierro-Azufre/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Mamíferos/genética , Mamíferos/metabolismo , Ratones , Ratones Noqueados , Mutación , Estrés Oxidativo , Células Procariotas/metabolismo , Salmonella enterica/genética , Transfección , Frataxina
13.
Eur J Nutr ; 49(7): 417-27, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20221766

RESUMEN

PURPOSE: Both dietary fat and dietary sucrose are major components of Western diets that may differentially affect the risk for body mass gain, diabetes mellitus, and cardiovascular disease. METHODS: We have phenotypically analyzed mice with ubiquitously impaired expression of mitochondrial frataxin protein that were challenged with diets differing in macronutrient content, namely high-sucrose/low-fat and high-saturated fat/low-sugar diets. RESULTS: We find here that a high-sucrose/low-fat diet has especially detrimental effects in mice with impaired mitochondrial metabolism promoting several independent cardiovascular risk factors, including impaired glucose metabolism, fasting hyperinsulinemia, reduced glucose-stimulated insulin secretion, increased serum triglycerides, and elevated cholesterol levels due to increased expression of HMG-CoA reductase. In contrast, a high-saturated fat/low-sugar diet protects mice with impaired mitochondrial metabolism from diet-induced obesity by increasing total energy expenditure and increasing expression of ACAA2, a rate-limiting enzyme of mitochondrial beta-oxidation, whereas no concomitant improvement of glucose metabolism was observed. CONCLUSIONS: Taken together, our results suggest that mitochondrial dysfunction may cause sucrose to become a multifunctional cardiovascular risk factor, whereas low-sugar diets high in saturated fat may prevent weight gain without improving glucose metabolism.


Asunto(s)
Enfermedades Cardiovasculares/dietoterapia , Grasas de la Dieta/sangre , Sacarosa en la Dieta/sangre , Glucosa/metabolismo , Mitocondrias/metabolismo , Animales , Sistema Cardiovascular/metabolismo , Colesterol/sangre , Colesterol/metabolismo , Diabetes Mellitus/dietoterapia , Dieta con Restricción de Grasas , Grasas de la Dieta/metabolismo , Sacarosa en la Dieta/metabolismo , Técnicas de Silenciamiento del Gen , Insulina/metabolismo , Secreción de Insulina , Proteínas de Unión a Hierro/metabolismo , Masculino , Ratones , Obesidad/dietoterapia , Oxidación-Reducción , Factores de Riesgo , Triglicéridos/sangre , Triglicéridos/metabolismo , Aumento de Peso , Frataxina
14.
PLoS One ; 2(10): e1013, 2007 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-17925861

RESUMEN

The Cre/loxP-system has become the system of choice for the generation of conditional so-called knockout mouse strains, i.e. the tissue-specific disruption of expression of a certain target gene. We here report the loss of expression of Cre recombinase in a transgenic mouse strain with increasing number of generations. This eventually led to the complete abrogation of gene expression of the inserted Cre cDNA while still being detectable at the genomic level. Conversely, loss of Cre expression caused an incomplete or even complete lack of disruption for the protein under investigation. As Cre expression in the tissue of interest in most cases cannot be addressed in vivo during the course of a study, our findings implicate the possibility that individual tail-biopsy genotypes may not necessarily indicate the presence or absence of gene disruption. This indicates that sustained post hoc analyses in regards to efficacy of disruption for every single study group member may be required.


Asunto(s)
Regulación de la Expresión Génica , Genotipo , Integrasas/genética , Transgenes , Animales , Biopsia , ADN Complementario/metabolismo , Genes Reporteros , Técnicas Genéticas , Ratones , Ratones Transgénicos , Fenotipo , Recombinación Genética , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN , Cola (estructura animal)
15.
Eur J Nutr ; 46(7): 397-405, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17882348

RESUMEN

BACKGROUND: Conflicting evidence suggests a possible role for vitamin E in mammalian glucose metabolism and the protection from type 2 diabetes. The alpha-tocopherol transfer protein (alpha-TTP) mediates the transfer of alpha-tocopherol (alpha-TOH) from hepatocytes to very-low-density lipoproteins, thereby controlling plasma levels of alpha-TOH. AIM OF THE STUDY: The aim of this study was to investigate the putative impact of alpha-TTP knock-out on glucose metabolism in mice. METHODS: Mice deficient for alpha-TTP and wild-type control littermates were fed a diet containing 200 mg alpha-tocopheryl acetate per kg to ameliorate alpha-TOH deficiency in knock-out mice. We investigated fasting and postprandial plasma glucose, insulin and triglyceride levels of both groups of mice at different ages. All genotypes and age groups were further subjected to glucose and insulin tolerance tests, and number of insulin-producing islets of Langerhans were determined. RESULTS: Plasma alpha-TOH levels of knock-out mice were 34% the levels of wild-type controls: Any signs of alpha-TOH deficiency were absent at any age. Unexpectedly, serum glucose levels both in the fasted and in the fed state were lower in alpha-TTP-deficient mice at any age. Removal rates for intraperitoneally injected glucose were found to be significantly increased in young alpha-TTP-deficient mice. This improved glucose tolerance was caused by increased insulin secretion in response to an intraperitoneal glucose challenge due to an increased number of pancreatic islets, as well as by increased sensitivity to intraperitoneally injected insulin, both significantly promoting glucose metabolism in alpha-TTP-deficient mice. CONCLUSIONS: Our findings suggest that alpha-TTP-deficiency in states of alpha-TOH supplementation unexpectedly promotes glucose tolerance in mice due to both increased insulin secretion and insulin action, suggesting differential roles of alpha-TTP and alpha-TOH in the pathogenesis of type 2 diabetes mellitus.


Asunto(s)
Proteínas Portadoras/fisiología , Glucosa/metabolismo , Insulina/sangre , Triglicéridos/sangre , alfa-Tocoferol/sangre , Factores de Edad , Animales , Proteínas Portadoras/genética , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/prevención & control , Ayuno/sangre , Femenino , Prueba de Tolerancia a la Glucosa , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Periodo Posprandial , Deficiencia de Vitamina E/complicaciones , alfa-Tocoferol/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...