Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(32): e2309967120, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37523551

RESUMEN

Body fat distribution is a heritable risk factor for cardiovascular and metabolic disease. In humans, rare Inhibin beta E (INHBE, activin E) loss-of-function variants are associated with a lower waist-to-hip ratio and protection from type 2 diabetes. Hepatic fatty acid sensing promotes INHBE expression during fasting and in obese individuals, yet it is unclear how the hepatokine activin E governs body shape and energy metabolism. Here, we uncover activin E as a regulator of adipose energy storage. By suppressing ß-agonist-induced lipolysis, activin E promotes fat accumulation and adipocyte hypertrophy and contributes to adipose dysfunction in mice. Mechanistically, we demonstrate that activin E elicits its effect on adipose tissue through ACVR1C, activating SMAD2/3 signaling and suppressing PPARG target genes. Conversely, loss of activin E or ACVR1C in mice increases fat utilization, lowers adiposity, and drives PPARG-regulated gene signatures indicative of healthy adipose function. Our studies identify activin E-ACVR1C as a metabolic rheostat promoting liver-adipose cross talk to restrain excessive fat breakdown and preserve fat mass during prolonged fasting, a mechanism that is maladaptive in obese individuals.


Asunto(s)
Diabetes Mellitus Tipo 2 , Lipólisis , Humanos , Ratones , Animales , Activinas/metabolismo , Adiposidad/genética , Diabetes Mellitus Tipo 2/metabolismo , PPAR gamma/metabolismo , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo
2.
Proc Natl Acad Sci U S A ; 112(6): 1845-9, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25624481

RESUMEN

G protein-coupled receptor 17 (GPR17) was recently reported to be a Foxo1 target in agouti-related peptide (AGRP) neurons. Intracerebroventricular injection of GPR17 agonists induced food intake, whereas administration of an antagonist to the receptor reduced feeding. These data lead to the conclusion that pharmacological modulation of GPR17 has therapeutic potential to treat obesity. Here we report that mice deficient in Gpr17 (Gpr17(-/-)) have similar food intake and body weight compared with their wild-type littermates. Gpr17(-/-) mice have normal hypothalamic Agrp mRNA expression, AGRP plasma levels, and metabolic rate. GPR17 deficiency in mice did not affect glucose homeostasis or prevent fat-induced insulin resistance. These data do not support a role for GPR17 in the control of food intake, body weight, or glycemic control.


Asunto(s)
Ingestión de Alimentos/genética , Glucosa/metabolismo , Proteínas del Tejido Nervioso/genética , Receptores Acoplados a Proteínas G/genética , Proteína Relacionada con Agouti/metabolismo , Análisis de Varianza , Animales , Secuencia de Bases , Composición Corporal/efectos de los fármacos , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Neuronas/metabolismo , Análisis de Secuencia de ARN , Factores de Tiempo , Microtomografía por Rayos X
3.
Stem Cell Reports ; 18(1): 394-409, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36525967

RESUMEN

Rats were more frequently used than mice to model human disease before mouse embryonic stem cells (mESCs) revolutionized genetic engineering in mice. Rat ESCs (rESCs) were first reported over 10 years ago, yet they are not as frequently used as mESCs. CRISPR-based gene editing in zygotes is widely used in rats but is limited by the difficulty of inserting or replacing DNA sequences larger than about 10 kb. We report here the generation of germline-competent rESC lines from several rat strains. These rESC lines maintain their potential for germline transmission after serial targeting with bacterial artificial chromosome (BAC)-based targeting vectors, and CRISPR-Cas9 cutting can increase targeting efficiency. Using these methods, we have successfully replaced entire rat genes spanning up to 101 kb with the human ortholog.


Asunto(s)
Células Madre Embrionarias , Degeneración Retiniana , Humanos , Ratas , Animales , Ratones , Edición Génica , Ingeniería Genética , Sistemas CRISPR-Cas/genética
4.
Sci Transl Med ; 15(723): eadd4897, 2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-37992152

RESUMEN

Deficiency in the adipose-derived hormone leptin or leptin receptor signaling causes class 3 obesity in individuals with genetic loss-of-function mutations in leptin or its receptor LEPR and metabolic and liver disease in individuals with hypoleptinemia secondary to lipoatrophy such as in individuals with generalized lipodystrophy. Therapies that restore leptin-LEPR signaling may resolve these metabolic sequelae. We developed a fully human monoclonal antibody (mAb), REGN4461 (mibavademab), that activates the human LEPR in the absence or presence of leptin. In obese leptin knockout mice, REGN4461 normalized body weight, food intake, blood glucose, and insulin sensitivity. In a mouse model of generalized lipodystrophy, REGN4461 alleviated hyperphagia, hyperglycemia, insulin resistance, dyslipidemia, and hepatic steatosis. In a phase 1, randomized, double-blind, placebo-controlled two-part study, REGN4461 was well tolerated with an acceptable safety profile. Treatment of individuals with overweight or obesity with REGN4461 decreased body weight over 12 weeks in those with low circulating leptin concentrations (<8 ng/ml) but had no effect on body weight in individuals with higher baseline leptin. Furthermore, compassionate-use treatment of a single patient with atypical partial lipodystrophy and a history of undetectable leptin concentrations associated with neutralizing antibodies to metreleptin was associated with noteable improvements in circulating triglycerides and hepatic steatosis. Collectively, these translational data unveil an agonist LEPR mAb that may provide clinical benefit in disorders associated with relatively low leptin concentrations.


Asunto(s)
Resistencia a la Insulina , Lipodistrofia Generalizada Congénita , Animales , Ratones , Humanos , Leptina/uso terapéutico , Ensayos de Uso Compasivo , Receptores de Leptina/metabolismo , Lipodistrofia Generalizada Congénita/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Anticuerpos/uso terapéutico , Peso Corporal
5.
Am J Physiol Endocrinol Metab ; 302(8): E987-91, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22318949

RESUMEN

To discover hypothalamic genes that might play a role in regulating energy balance, we carried out a microarray screen for genes induced by a 48-h fast in male C57Bl/6J mouse hypothalamus. One such gene was Fkbp51 (FK506 binding protein 5; Locus NP_034350). The product of this gene is of interest because it blocks glucocorticoid action, suggesting that fasting-induced elevation of this gene in the hypothalamus may reduce glucocorticoid negative feedback, leading to elevated glucocorticoid levels, thus promoting obese phenotypes. Subsequent analysis demonstrated that a 48-h fast induces Fkbp51 in ventromedial, paraventricular, and arcuate hypothalamic nuclei of mice and rats. To assess if hypothalamic Fkbp51 promotes obesity, the gene was transferred to the hypothalamus via an adeno-associated virus vector. Within 2 wk following Fkbp51 overexpression, mice on a high-fat diet exhibited elevated body weight, without hyperphagia, relative to mice receiving the control mCherry vector. Body weight remained elevated for more than 8 wk and was associated with elevated corticosterone and impaired glucose tolerance. These studies suggest that elevated hypothalamic Fkbp51 promotes obese phenotypes.


Asunto(s)
Ayuno/metabolismo , Hipotálamo/metabolismo , Obesidad/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Regulación hacia Arriba , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Corticosterona/sangre , Ingestión de Energía , Perfilación de la Expresión Génica , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/sangre , Análisis de Secuencia por Matrices de Oligonucleótidos , Núcleo Hipotalámico Paraventricular/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/genética , Núcleo Hipotalámico Ventromedial/metabolismo , Aumento de Peso
6.
J Bone Miner Res ; 36(4): 739-756, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33249643

RESUMEN

Osteogenesis imperfecta (OI) is a genetic connective tissue disorder characterized by compromised skeletal integrity, altered microarchitecture, and bone fragility. Current OI treatment strategies focus on bone antiresorptives and surgical intervention with limited effectiveness, and thus identifying alternative therapeutic options remains critical. Muscle is an important stimulus for bone formation. Myostatin, a TGF-ß superfamily myokine, acts through ActRIIB to negatively regulate muscle growth. Recent studies demonstrated the potential benefit of myostatin inhibition with the soluble ActRIIB fusion protein on skeletal properties, although various OI mouse models exhibited variable skeletal responses. The genetic and clinical heterogeneity associated with OI, the lack of specificity of the ActRIIB decoy molecule for myostatin alone, and adverse events in human clinical trials further the need to clarify myostatin's therapeutic potential and role in skeletal integrity. In this study, we determined musculoskeletal outcomes of genetic myostatin deficiency and postnatal pharmacological myostatin inhibition by a monoclonal anti-myostatin antibody (Regn647) in the G610C mouse, a model of mild-moderate type I/IV human OI. In the postnatal study, 5-week-old wild-type and +/G610C male and female littermates were treated with Regn647 or a control antibody for 11 weeks or for 7 weeks followed by a 4-week treatment holiday. Inhibition of myostatin, whether genetically or pharmacologically, increased muscle mass regardless of OI genotype, although to varying degrees. Genetic myostatin deficiency increased hindlimb muscle weights by 6.9% to 34.4%, whereas pharmacological inhibition increased them by 13.5% to 29.6%. Female +/mstn +/G610C (Dbl.Het) mice tended to have similar trabecular and cortical bone parameters as Wt showing reversal of +/G610C characteristics but with minimal effect of +/mstn occurring in male mice. Pharmacologic myostatin inhibition failed to improve skeletal bone properties of male or female +/G610C mice, although skeletal microarchitectural and biomechanical improvements were observed in male wild-type mice. Four-week treatment holiday did not alter skeletal outcomes. © 2020 American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Osteogénesis Imperfecta , Animales , Huesos , Colágeno Tipo I , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Miostatina/genética , Osteogénesis , Osteogénesis Imperfecta/tratamiento farmacológico , Osteogénesis Imperfecta/genética
7.
Endocrinology ; 148(4): 1928-32, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17218412

RESUMEN

Because appetite, hypothalamic gene expression, reproductive function, and adrenal function are highly sensitive to acute changes in plasma glucose levels, it has been hypothesized hypothalamic neurons sensitive to glucose play a role in regulating these functions. To assess this hypothesis, we examined these neuronendocrine functions in mice in which the glucokinase gene, which plays an essential role in neuroendocrine glucose sensing, has been ablated. Haploinsufficiency in heterozygous glucokinase knockout mice produced effects similar to those produced by hypoglycemia: impaired reproductive function, elevated plasma corticosterone, increased food intake, and hypothalamic gene expression similar to that observed in fasted or leptin-deficient obese mice (increased hypothalamic neuropeptide Y mRNA and reduced hypothalamic proopiomelanocortin mRNA). Plasma glucose was elevated 2-fold in glucokinase knockout mice, consistent with a maturity-onset diabetes of the young phenotype, but plasma insulin and leptin levels were normal. These data support the hypothesis that glucokinase plays a key role in the neuroendocrine regulation of metabolic economy.


Asunto(s)
Ingestión de Alimentos/genética , Glucocorticoides/metabolismo , Glucoquinasa/fisiología , Hipotálamo/metabolismo , Reproducción/genética , Animales , Femenino , Fertilidad/genética , Regulación de la Expresión Génica , Glucoquinasa/genética , Hiperglucemia/genética , Insulina/sangre , Insulina/genética , Leptina/sangre , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas
8.
Nat Commun ; 8: 15153, 2017 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-28452368

RESUMEN

Growth and differentiation factor 8 (GDF8) is a TGF-ß superfamily member, and negative regulator of skeletal muscle mass. GDF8 inhibition results in prominent muscle growth in mice, but less impressive hypertrophy in primates, including man. Broad TGF-ß inhibition suggests another family member negatively regulates muscle mass, and its blockade enhances muscle growth seen with GDF8-specific inhibition. Here we show that activin A is the long-sought second negative muscle regulator. Activin A specific inhibition, on top of GDF8 inhibition, leads to pronounced muscle hypertrophy and force production in mice and monkeys. Inhibition of these two ligands mimics the hypertrophy seen with broad TGF-ß blockers, while avoiding the adverse effects due to inhibition of multiple family members. Altogether, we identify activin A as a second negative regulator of muscle mass, and suggest that inhibition of both ligands provides a preferred therapeutic approach, which maximizes the benefit:risk ratio for muscle diseases in man.


Asunto(s)
Activinas/metabolismo , Hipertrofia/patología , Hipotonía Muscular/patología , Músculo Esquelético/crecimiento & desarrollo , Miostatina/metabolismo , Receptores de Activinas Tipo II/metabolismo , Activinas/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/farmacología , Índice de Masa Corporal , Dexametasona/farmacología , Humanos , Contracción Isométrica/fisiología , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Músculo Esquelético/fisiología , Miostatina/antagonistas & inhibidores , Ratas
9.
Diabetes ; 54(4): 952-8, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15793232

RESUMEN

The robust neuroendocrine counterregulatory responses induced by hypoglycemia protect the brain by restoring plasma glucose, but little is known about molecular responses to hypoglycemia that may also be neuroprotective. To clarify these mechanisms, we examined gene expression in hypothalamus, cortex, and liver 3 h after induction of mild hypoglycemia by a single injection of insulin, using cDNA microarray analysis and quantitative real-time PCR. Real-time PCR corroborated the induction of six genes (angiotensinogen, GLUT-1, inhibitor of kappaB, inhibitor of DNA binding 1 [ID-1], Ubp41, and mitogen-activated protein kinase phosphatase-1 [MKP-1]) by insulin-induced hypoglycemia in the hypothalamus: five of these six genes in cortex and three (GLUT-1, angiotensinogen, and MKP-1) in liver. The induction was due to hypoglycemia and not hyperinsulinemia, since fasting (characterized by low insulin and glucose) also induced these genes. Four of these genes (angiotensinogen, GLUT-1, ID-1, and MKP-1) have been implicated in enhancement of glucose availability, which could plausibly serve a neuroprotective role during acute hypoglycemia but, if persistent, could also cause glucose-sensing mechanisms to overestimate plasma glucose levels, potentially causing hypoglycemia-induced counterregulatory failure. Although using cDNA microarrays with more genes, or microdissection, would presumably reveal further responses to hypoglycemia, these hypoglycemia-induced genes represent useful markers to assess molecular mechanisms mediating cellular responses to hypoglycemia.


Asunto(s)
Encéfalo/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hipoglucemia/inducido químicamente , Hipoglucemia/metabolismo , Insulina/farmacología , Hígado/metabolismo , Animales , Regulación hacia Abajo , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Regulación hacia Arriba
10.
Physiol Behav ; 85(1): 3-23, 2005 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-15924903

RESUMEN

Since nutrition-sensitive feedback signals normally act to maintain relatively stable levels of both available and stored nutritional resources, failure in one or more of these feedback signals could plausibly lead to obese phenotypes. The glucostatic hypothesis in its original form posited that glucose serves as a physiological satiety factor (in the sense that post-prandial increases in plasma glucose cause meal termination), but in this form the hypothesis has been difficult to prove, and, especially since the discovery of leptin, the glucostatic hypothesis has largely been abandoned. Nevertheless, reduction of plasma glucose levels or glucose signaling produces a profile of neuroendocrine responses similar to those produced by leptin deficiency. Since leptin is not a physiological satiety factor (because it does not increase before meal termination), yet leptin deficiency causes obesity, we suggest that the glucostatic hypothesis be re-formulated without reference to satiety (i.e., short-term effects on food intake). Instead we argue that like leptin signaling, glucose signaling regulates long-term energy balance, in part by regulating metabolic rate but also by chronically regulating food intake. We further speculate that high-fat diets produce obesity in part because carbohydrates are, per calorie, more effective than lipids to reduce food intake and increase metabolic rate. In support of this glucoadipostatic hypothesis, the 5 present review examines evidence that obesity and the metabolic syndrome may be due to reduction in neuroendocrine sensitivity to glucose leading to increased metabolic efficiency.


Asunto(s)
Metabolismo Energético , Glucosa/metabolismo , Obesidad/etiología , Transducción de Señal , Adenilato Quinasa/metabolismo , Animales , Glucemia/metabolismo , Dieta , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiopatología , Insulina/metabolismo , Leptina/metabolismo , Obesidad/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proopiomelanocortina/fisiología , Respuesta de Saciedad/fisiología , Termogénesis/fisiología
11.
Diabetes ; 64(10): 3564-72, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26130763

RESUMEN

The objective of this study was to determine whether the sodium-glucose transporter SGLT1 in the ventromedial hypothalamus (VMH) plays a role in glucose sensing and in regulating the counterregulatory response to hypoglycemia, and if so, whether knockdown of in the VMH can improve counterregulatory responses to hypoglycemia in diabetic rats or rats exposed to recurrent bouts of hypoglycemia (RH). Normal Sprague-Dawley rats as well as RH or streptozotocin (STZ)-diabetic rats received bilateral VMH microinjections of an adenoassociated viral vector containing either the SGLT1 short hairpin RNA (shRNA) or a scrambled RNA sequence. Subsequently, these rats underwent a hypoglycemic clamp to assess hormone responses. In a subgroup of rats, glucose kinetics was determined using tritiated glucose. The shRNA reduced VMH SGLT1 expression by 53% in nondiabetic rats, and this augmented glucagon and epinephrine responses and hepatic glucose production during hypoglycemia. Similarly, SGLT1 knockdown improved the glucagon and epinephrine responses in RH rats and restored the impaired epinephrine response to hypoglycemia in STZ-diabetic animals. These findings suggest that SGLT1 in the VMH plays a significant role in the detection and activation of counterregulatory responses to hypoglycemia. Inhibition of SGLT1 may offer a potential therapeutic target to diminish the risk of hypoglycemia in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Regulación de la Expresión Génica/fisiología , Hipoglucemia/metabolismo , ARN Mensajero/metabolismo , Transportador 1 de Sodio-Glucosa/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo , Animales , Glucemia , Masculino , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Transportador 1 de Sodio-Glucosa/genética
12.
Skelet Muscle ; 5: 34, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26457176

RESUMEN

BACKGROUND: Loss of skeletal muscle mass and function in humans is associated with significant morbidity and mortality. The role of myostatin as a key negative regulator of skeletal muscle mass and function has supported the concept that inactivation of myostatin could be a useful approach for treating muscle wasting diseases. METHODS: We generated a myostatin monoclonal blocking antibody (REGN1033) and characterized its effects in vitro using surface plasmon resonance biacore and cell-based Smad2/3 signaling assays. REGN1033 was tested in mice for the ability to induce skeletal muscle hypertrophy and prevent atrophy induced by immobilization, hindlimb suspension, or dexamethasone. The effect of REGN1033 on exercise training was tested in aged mice. Messenger RNA sequencing, immunohistochemistry, and ex vivo force measurements were performed on skeletal muscle samples from REGN1033-treated mice. RESULTS: The human monoclonal antibody REGN1033 is a specific and potent myostatin antagonist. Chronic treatment of mice with REGN1033 increased muscle fiber size, muscle mass, and force production. REGN1033 prevented the loss of muscle mass induced by immobilization, glucocorticoid treatment, or hindlimb unweighting and increased the gain of muscle mass during recovery from pre-existing atrophy. In aged mice, REGN1033 increased muscle mass and strength and improved physical performance during treadmill exercise. CONCLUSIONS: We show that specific myostatin antagonism with the human antibody REGN1033 enhanced muscle mass and function in young and aged mice and had beneficial effects in models of skeletal muscle atrophy.

13.
Endocrinology ; 156(12): 4502-10, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26406932

RESUMEN

Secreted frizzled-related protein 4 (SFRP4) is an extracellular regulator of the wingless-type mouse mammary tumor virus integration site family (WNT) pathway. SFRP4 has been implicated in adipocyte dysfunction, obesity, insulin resistance, and impaired insulin secretion in patients with type 2 diabetes. However, the exact role of SFRP4 in regulating whole-body metabolism and glucose homeostasis is unknown. We show here that male Sfrp4(-/-) mice have increased spine length and gain more weight when fed a high-fat diet. The body composition and body mass per spine length of diet-induced obese Sfrp4(-/-) mice is similar to wild-type littermates, suggesting that the increase in body weight can be accounted for by their longer body size. The diet-induced obese Sfrp4(-/-) mice have reduced energy expenditure, food intake, and bone mineral density. Sfrp4(-/-) mice have normal glucose and insulin tolerance and ß-cell mass. Diet-induced obese Sfrp4(-/-) and control mice show similar impairments of glucose tolerance and a 5-fold compensatory expansion of their ß-cell mass. In summary, our data suggest that loss of SFRP4 alters body length and bone mineral density as well as energy expenditure and food intake. However, SFRP4 does not control glucose homeostasis and ß-cell mass in mice.


Asunto(s)
Tamaño Corporal/genética , Densidad Ósea/genética , Dieta Alta en Grasa , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Células Secretoras de Insulina/metabolismo , Obesidad , Proteínas Proto-Oncogénicas/genética , Animales , Glucemia/metabolismo , Composición Corporal/genética , Conducta Alimentaria , Técnicas de Sustitución del Gen , Prueba de Tolerancia a la Glucosa , Células HEK293 , Homeostasis/genética , Humanos , Insulina/metabolismo , Masculino , Ratones , Ratones Noqueados , Vía de Señalización Wnt , Microtomografía por Rayos X
14.
BMC Neurosci ; 3: 18, 2002 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-12423556

RESUMEN

BACKGROUND: Several lines of evidence strongly suggest that agouti-related peptide (AGRP) plays a key role in the regulation of metabolic function but ablation of the AGRP gene has no apparent effect on metabolic function. Since specific pharmacological antagonists of AGRP do not presently exist, we assessed if reduction of hypothalamic AGRP mRNA by RNA interference (RNAI) would influence metabolic function, an outcome suggesting that pharmacological antagonists might constitute useful reagents to treat obesity. RESULTS: The RNAI protocol specifically reduced hypothalamic expression of AGRP mRNA by 50% and resulted in reduction of AGRP peptide immunoreactivity. Physiologically, the reduction in AGRP levels was associated with increased metabolic rate and reduced body weight without changes in food intake. CONCLUSION: AGRP can function to increase body weight and reduce metabolic rate without influencing food intake. The present study demonstrates that RNAI protocols can be used to assess physiological function of neuronal genes in vivo.


Asunto(s)
Metabolismo Basal/fisiología , Peso Corporal/fisiología , Ingestión de Alimentos , Hipotálamo/metabolismo , Proteínas/fisiología , ARN Interferente Pequeño/farmacología , Proteína Relacionada con Agouti , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Basal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Calorimetría Indirecta , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Ingestión de Alimentos/efectos de los fármacos , Proteínas Fluorescentes Verdes , Hipotálamo/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular , Proteínas Luminiscentes/antagonistas & inhibidores , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos C57BL , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Proteínas/antagonistas & inhibidores , ARN Mensajero/metabolismo
15.
Brain Res ; 961(2): 255-60, 2003 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-12531492

RESUMEN

The peroxisome proliferator activated receptor coactivator 1 alpha (PGC-1alpha) is a nuclear transcriptional coactivator that is expressed in brown adipose tissue, brain, heart and kidney as well as cold-exposed skeletal muscle. In liver, white and brown adipose tissue, PGC-1alpha expression is regulated in a manner suggesting a role in energy homeostasis. To characterize PGC-1alpha expression in the rodent brain and to determine brain PGC-1alpha regulation, we used in situ hybridization histochemistry in C57Bl/6J mice and Sprague-Dawley rats. We found that PGC-1alpha is widely expressed in brain areas, including in the olfactory bulb, cerebral cortex, the diagonal band of Broca, the medial septal nucleus, reticular thalamic nucleus, the striatum and globus pallidus, the hippocampus, the substantia nigra, the mesencephalic nucleus of the trigeminal nerve, the cochlear nucleus and the superior olivary complex. In contrast, PGC-1alpha expression was absent in the hypothalamus. To evaluate PGC-1alpha expression under different physiologic states in these various brain areas, we examined expression with fasting, leptin treatment and cold exposure (4 h at 4 degrees C) and found no change, nor was expression changed in the brain of the leptin-deficient ob/ob mice and the hyperleptinemic UCP-DTA mice. Hence, PGC-1alpha is widely expressed in the rodent brain, but is not regulated by states of caloric deficiency, leptin, obesity or cold exposure. Its functional role in the brain requires further study.


Asunto(s)
Encéfalo/metabolismo , Frío , Ayuno/metabolismo , Leptina/metabolismo , Factores de Transcripción/metabolismo , Animales , Hibridación in Situ , Leptina/administración & dosificación , Leptina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Obesidad/metabolismo , Ratas , Ratas Sprague-Dawley , Temperatura
16.
J Child Neurol ; 28(8): 1009-14, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23680948

RESUMEN

Accumulating evidence suggests that low-carbohydrate, high-fat diets are safe and effective to reduce glycemia in diabetic patients without producing significant cardiovascular risks. Most of these studies have been carried out specifically restricting carbohydrates, which tends to lead to increased protein intake, thus reducing the ketosis. However, diets that limit protein as well as carbohydrates, entailing a composition very high in fat, appear even more effective to reduce glucose and whole-body glucose metabolism in humans. In animal models, low-carbohydrate, high-protein diets do not produce ketosis or reduce glycemia but rather cause obesity. However, limiting both protein and carbohydrates as in a classic ketogenic diet remarkably reduces blood glucose in animal models of type 1 and type 2 diabetes and reverses diabetic nephropathy. Future studies should assess if ketogenic diets would be effective to reverse diabetic complications in humans.


Asunto(s)
Complicaciones de la Diabetes/dietoterapia , Dieta Cetogénica/métodos , Obesidad/dietoterapia , Animales , Glucemia/metabolismo , Modelos Animales de Enfermedad , Metabolismo Energético , Humanos , Obesidad/sangre
17.
Diabetes ; 60(1): 39-46, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20811039

RESUMEN

OBJECTIVE: Hypoglycemia-associated autonomic failure (HAAF) constitutes one of the main clinical obstacles to optimum treatment of type 1 diabetes. Neurons in the ventromedial hypothalamus are thought to mediate counterregulatory responses to hypoglycemia. We have previously hypothesized that hypoglycemia-induced hypothalamic angiotensin might contribute to HAAF, suggesting that the angiotensin blocker valsartan might prevent HAAF. On the other hand, clinical studies have demonstrated that the opioid receptor blocker naloxone ameliorates HAAF. The goal of this study was to generate novel hypothalamic markers of hypoglycemia and use them to assess mechanisms mediating HAAF and its reversal. RESEARCH DESIGN AND METHODS: Quantitative PCR was used to validate a novel panel of hypothalamic genes regulated by hypoglycemia. Mice were exposed to one or five episodes of insulin-induced hypoglycemia, with or without concurrent exposure to valsartan or naloxone. Corticosterone, glucagon, epinephrine, and hypothalamic gene expression were assessed after the final episode of hypoglycemia. RESULTS: A subset of hypothalamic genes regulated acutely by hypoglycemia failed to respond after repetitive hypoglycemia. Responsiveness of a subset of these genes was preserved by naloxone but not valsartan. Notably, hypothalamic expression of four genes, including pyruvate dehydrogenase kinase 4 and glycerol 3-phosphate dehydrogenase 1, was acutely induced by a single episode of hypoglycemia, but not after antecedent hypoglycemia; naloxone treatment prevented this failure. Similarly, carnitine palmitoyltransferase-1 was inhibited after repetitive hypoglycemia, and this inhibition was prevented by naloxone. Repetitive hypoglycemia also caused a loss of hypoglycemia-induced elevation of glucocorticoid secretion, a failure prevented by naloxone but not valsartan. CONCLUSIONS: Based on these observations we speculate that acute hypoglycemia induces reprogramming of hypothalamic metabolism away from glycolysis toward ß-oxidation, HAAF is associated with a reversal of this reprogramming, and naloxone preserves some responses to hypoglycemia by preventing this reversal.


Asunto(s)
Hipoglucemia/complicaciones , Naloxona/farmacología , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/genética , Animales , Antihipertensivos/farmacología , Proteínas Reguladoras de la Apoptosis , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Proteínas Portadoras , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Transportador de Glucosa de Tipo 2/genética , Glicerolfosfato Deshidrogenasa/genética , Homeostasis/efectos de los fármacos , Hipoglucemia/inducido químicamente , Hipoglucemia/etiología , Hipoglucemia/genética , Insulina/farmacología , Proteínas de la Membrana/genética , Ratones , Naloxona/uso terapéutico , Antagonistas de Narcóticos/farmacología , Perilipina-4 , Proteínas Serina-Treonina Quinasas/genética , Insuficiencia Autonómica Pura/etiología , Insuficiencia Autonómica Pura/prevención & control , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Proteínas Represoras , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tetrazoles/farmacología , Factores de Transcripción/genética , Valina/análogos & derivados , Valina/farmacología , Valsartán
18.
PLoS One ; 6(4): e18604, 2011 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-21533091

RESUMEN

Intensive insulin therapy and protein restriction delay the development of nephropathy in a variety of conditions, but few interventions are known to reverse nephropathy. Having recently observed that the ketone 3-beta-hydroxybutyric acid (3-OHB) reduces molecular responses to glucose, we hypothesized that a ketogenic diet, which produces prolonged elevation of 3-OHB, may reverse pathological processes caused by diabetes. To address this hypothesis, we assessed if prolonged maintenance on a ketogenic diet would reverse nephropathy produced by diabetes. In mouse models for both Type 1 (Akita) and Type 2 (db/db) diabetes, diabetic nephropathy (as indicated by albuminuria) was allowed to develop, then half the mice were switched to a ketogenic diet. After 8 weeks on the diet, mice were sacrificed to assess gene expression and histology. Diabetic nephropathy, as indicated by albumin/creatinine ratios as well as expression of stress-induced genes, was completely reversed by 2 months maintenance on a ketogenic diet. However, histological evidence of nephropathy was only partly reversed. These studies demonstrate that diabetic nephropathy can be reversed by a relatively simple dietary intervention. Whether reduced glucose metabolism mediates the protective effects of the ketogenic diet remains to be determined.


Asunto(s)
Nefropatías Diabéticas/dietoterapia , Dieta Cetogénica , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
19.
Endocrinology ; 151(11): 5206-17, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20881243

RESUMEN

Nutrient-sensitive hypothalamic neurons regulate energy balance and glucose homeostasis, but the molecular mechanisms mediating hypothalamic responses to nutritional state remain incompletely characterized. To address these mechanisms, the present studies used quantitative PCR to characterize the expression of a panel of genes the hypothalamic expression by nutritional status of which had been suggested by DNA microarray studies. Although these genes regulate a variety of function, the most prominent set regulate intermediary metabolism, and the overall pattern clearly indicated that a 48-h fast produced a metabolic reprogramming away from glucose metabolism and toward the utilization of alternative fuels, particularly lipid metabolism. This general reprogramming of intermediary metabolism by fasting was observed both in cortex and hypothalamus but most prominently in hypothalamus. The effect of fasting on the expression of these genes may be mediated by reduction in plasma glucose or glucose metabolism, rather than leptin, because they were generally recapitulated by hypoglycemia even in the presence of elevated insulin and in vitro by low glucose but were not recapitulated in ob/ob mice. These studies suggest that fasting reduces glucose metabolism and thus minimizes the production of hypothalamic malonyl-coenzyme A. However, because the reprogramming of glucose metabolism by fasting was also observed in cortex, this apparent substrate competition may mediate more general responses to nutritional deprivation, including those responsible for the protective effects of dietary restriction. The present studies also provide a large panel of novel glucose-regulated genes that can be used as markers of glucose action to address mechanisms mediating hypothalamic responses to nutritional state.


Asunto(s)
Ayuno/metabolismo , Glucólisis/fisiología , Hipotálamo/metabolismo , Metabolismo de los Lípidos/fisiología , Neuronas/metabolismo , Estado Nutricional/fisiología , Análisis de Varianza , Animales , Corteza Cerebral/metabolismo , Expresión Génica , Insulina/metabolismo , Leptina/metabolismo , Masculino , Ratones , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Appetite ; 48(2): 135-8, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17141367

RESUMEN

High-fat diets produce obesity in part because, per calorie, glucose produces greater post-prandial thermogenesis than lipids, an effect probably mediated by glucose-sensing neurons. A very low-carbohydrate/high-fat/high-protein Atkins-type diet produces obesity but is marginally ketogenic in mice. In contrast, high-sucrose/low-fat diets, and very low-carbohydrate/high-fat/low-protein (anti-epileptic) ketogenic diets reverse diet-induced obesity independent of caloric intake. We propose that a non-ketogenic high-fat diet reduces glucose metabolism and signaling in glucose-sensing neurons, thereby reducing post-prandial thermogenesis, and that a ketogenic high-fat diet does not reduce glucose signaling, thereby preventing and/or reversing obesity.


Asunto(s)
Dieta Baja en Carbohidratos , Dieta Reductora , Metabolismo Energético/fisiología , Obesidad/dietoterapia , Obesidad/etiología , Glucemia/metabolismo , Restricción Calórica , Dieta Baja en Carbohidratos/efectos adversos , Carbohidratos de la Dieta/administración & dosificación , Ingestión de Energía/fisiología , Humanos , Obesidad/epidemiología , Obesidad/metabolismo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA