Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38659949

RESUMEN

Background and Aims: The visceral organ-brain axis, mediated by vagal sensory neurons in the vagal nerve ganglion, is essential for maintaining various physiological functions. In this study, we investigated the impact of liver-projecting vagal sensory neurons on energy balance, hepatic steatosis, and anxiety-like behavior in mice under obesogenic conditions. Methods: We performed single-nucleus RNA sequencing of vagal sensory neurons innervating the liver. Based on our snRNA-Seq results, we used the Avil CreERT2 strain to identify vagal sensory neurons that innervate the liver. Results: A small subset of polymodal sensory neurons innervating the liver was located in the left and right ganglia, projecting centrally to the nucleus of the tractus solitarius, area postrema, and dorsal motor nucleus of the vagus, and peripherally to the periportal areas in the liver. Male and female control mice developed diet-induced obesity (DIO) during high-fat diet feeding. Deleting liver-projecting advillin-positive vagal sensory neurons prevented DIO in male and female mice, and these outcomes are associated with increased energy expenditure. Although males and females exhibited improved glucose homeostasis following disruption of liver-projecting vagal sensory neurons, only male mice displayed increased insulin sensitivity. The loss of liver-projecting vagal sensory neurons limited the progression of hepatic steatosis in male and female mice fed a steatogenic diet. Finally, mice lacking liver-innervating vagal sensory neurons exhibited less anxiety-like behavior compared to the control mice. Conclusions: The liver-brain axis contributes to the regulation of energy balance, glucose tolerance, hepatic steatosis, and anxiety-like behavior depending on the nutrient status in healthy and obesogenic conditions.

2.
Dev Cell ; 56(6): 747-760.e6, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33667344

RESUMEN

Loss of insulin-secreting pancreatic ß cells through apoptosis contributes to the progression of type 2 diabetes, but underlying mechanisms remain elusive. Here, we identify a pathway in which the cell death inhibitor ARC paradoxically becomes a killer during diabetes. While cytoplasmic ARC maintains ß cell viability and pancreatic architecture, a pool of ARC relocates to the nucleus to induce ß cell apoptosis in humans with diabetes and several pathophysiologically distinct mouse models. ß cell death results through the coordinate downregulation of serpins (serine protease inhibitors) not previously known to be synthesized and secreted by ß cells. Loss of the serpin α1-antitrypsin from the extracellular space unleashes elastase, triggering the disruption of ß cell anchorage and subsequent cell death. Administration of α1-antitrypsin to mice with diabetes prevents ß cell death and metabolic abnormalities. These data uncover a pathway for ß cell loss in type 2 diabetes and identify an FDA-approved drug that may impede progression of this syndrome.


Asunto(s)
Apoptosis , Núcleo Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/patología , Células Secretoras de Insulina/patología , Proteínas del Tejido Nervioso/metabolismo , alfa 1-Antitripsina/química , Animales , Proteínas Reguladoras de la Apoptosis/fisiología , Citoplasma/metabolismo , Proteínas del Citoesqueleto/genética , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/fisiología , Proteínas del Tejido Nervioso/genética , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo
3.
Nat Commun ; 11(1): 6295, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33293550

RESUMEN

The central melanocortin system plays a fundamental role in the control of feeding and body weight. Proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARC) also regulate overall glucose homeostasis via insulin-dependent and -independent pathways. Here, we report that a subset of ARC POMC neurons innervate the liver via preganglionic parasympathetic acetylcholine (ACh) neurons in the dorsal motor nucleus of the vagus (DMV). Optogenetic stimulation of this liver-projecting melanocortinergic pathway elevates blood glucose levels that is associated with increased expression of hepatic gluconeogenic enzymes in female and male mice. Pharmacological blockade and knockdown of the melanocortin-4 receptor gene in the DMV abolish this stimulation-induced effect. Activation of melanocortin-4 receptors inhibits DMV cholinergic neurons and optogenetic inhibition of liver-projecting parasympathetic cholinergic fibers increases blood glucose levels. This elevated blood glucose is not due to altered pancreatic hormone release. Interestingly, insulin-induced hypoglycemia increases ARC POMC neuron activity. Hence, this liver-projecting melanocortinergic circuit that we identified may play a critical role in the counterregulatory response to hypoglycemia.


Asunto(s)
Glucemia/metabolismo , Hipoglucemia/etiología , Hígado/inervación , Proopiomelanocortina/metabolismo , Nervio Vago/metabolismo , Acetilcolina/metabolismo , Potenciales de Acción/fisiología , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/metabolismo , Glucemia/análisis , Neuronas Colinérgicas/metabolismo , Corticosterona/sangre , Corticosterona/metabolismo , Modelos Animales de Enfermedad , Vías Eferentes/fisiología , Femenino , Técnicas de Silenciamiento del Gen , Glucagón/sangre , Glucagón/metabolismo , Gluconeogénesis/genética , Humanos , Hipoglucemia/sangre , Hipoglucemia/diagnóstico , Insulina/sangre , Insulina/metabolismo , Hígado/enzimología , Masculino , Ratones , Optogenética , ARN Mensajero/metabolismo , Receptor de Melanocortina Tipo 4/antagonistas & inhibidores , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Regulación hacia Arriba , Nervio Vago/citología
4.
Trends Endocrinol Metab ; 31(1): 46-52, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31629614

RESUMEN

Obesity is a neurological disorder that operates by favoring energy storage within adipose depots and increased caloric intake. Most cases of human obesity are acquired without any underlying genetic basis. Here, we suggest that obesity can impair the function of some hypothalamic neurons critical to body weight regulation. Genetic ablation of the retinoblastoma (Rb) gene within pro-opiomelanocortin (POMC) neurons leads to death of the neurons and subsequent obesity. The Rb protein (pRb), a key inhibitor of the cell cycle, can also be inactivated by cyclin dependent kinase (CDK)-mediated phosphorylation. Extensive development led to the production of FDA-approved CDK4/6 inhibitors. Based on our own results, we propose that maintaining or re-instating pRb function using CDK4/6 inhibitors are potentially effective treatments of diet-induced obesity (DIO).


Asunto(s)
Obesidad/metabolismo , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Humanos , Hipotálamo/metabolismo , Proopiomelanocortina/metabolismo , Retinoblastoma/metabolismo
5.
J Immunol ; 201(7): 1837-1841, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30135180

RESUMEN

Various malignancies are reproducibly cured in mouse models, but most cancer immunotherapies show objective responses in a fraction of treated patients. One reason for this disconnect may be the use of young, lean mice lacking immune-altering comorbidities present in cancer patients. Although many cancer patients are overweight or obese, the effect of obesity on antitumor immunity is understudied in preclinical tumor models. We examined the effect of obesity on two immunotherapeutic models: systemic anti-CTLA-4 mAb and intratumoral delivery of a TRAIL-encoding adenovirus plus CpG. Both therapies were effective in lean mice, but neither provided a survival benefit to diet-induced obese BALB/c mice. Interestingly, tumor-bearing leptin-deficient (ob/ob) obese BALB/c mice did respond to treatment. Moreover, reducing systemic leptin with soluble leptin receptor:Fc restored the antitumor response in diet-induced obese mice. These data demonstrate the potential of targeting leptin to improve tumor immunotherapy when immune-modulating comorbidities are present.


Asunto(s)
Adenocarcinoma/metabolismo , Envejecimiento/fisiología , Anticuerpos Monoclonales/uso terapéutico , Inmunoterapia/métodos , Neoplasias Renales/metabolismo , Leptina/metabolismo , Obesidad/metabolismo , Adenocarcinoma/terapia , Adenoviridae/genética , Animales , Antígeno CTLA-4/inmunología , Línea Celular Tumoral , Dieta , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad , Neoplasias Renales/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Obesidad/terapia , Oligodesoxirribonucleótidos/metabolismo , Receptores Fc/genética , Receptores Fc/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
6.
PLoS Biol ; 16(4): e2004399, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29689050

RESUMEN

Proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARC) respond to numerous hormonal and neural signals, resulting in changes in food intake. Here, we demonstrate that ARC POMC neurons express capsaicin-sensitive transient receptor potential vanilloid 1 receptor (TRPV1)-like receptors. To show expression of TRPV1-like receptors in ARC POMC neurons, we use single-cell reverse transcription-polymerase chain reaction (RT-PCR), immunohistochemistry, electrophysiology, TRPV1 knock-out (KO), and TRPV1-Cre knock-in mice. A small elevation of temperature in the physiological range is enough to depolarize ARC POMC neurons. This depolarization is blocked by the TRPV1 receptor antagonist and by Trpv1 gene knockdown. Capsaicin-induced activation reduces food intake that is abolished by a melanocortin receptor antagonist. To selectively stimulate TRPV1-like receptor-expressing ARC POMC neurons in the ARC, we generate an adeno-associated virus serotype 5 (AAV5) carrying a Cre-dependent channelrhodopsin-2 (ChR2)-enhanced yellow fluorescent protein (eYFP) expression cassette under the control of the two neuronal POMC enhancers (nPEs). Optogenetic stimulation of TRPV1-like receptor-expressing POMC neurons decreases food intake. Hypothalamic temperature is rapidly elevated and reaches to approximately 39 °C during treadmill running. This elevation is associated with a reduction in food intake. Knockdown of the Trpv1 gene exclusively in ARC POMC neurons blocks the feeding inhibition produced by increased hypothalamic temperature. Taken together, our findings identify a melanocortinergic circuit that links acute elevations in hypothalamic temperature with acute reductions in food intake.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Ingestión de Alimentos/genética , Neuronas/metabolismo , Proopiomelanocortina/genética , Canales Catiónicos TRPV/genética , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Capsaicina/farmacología , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Elementos de Facilitación Genéticos , Femenino , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Genes Reporteros , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Transgénicos , Neuronas/citología , Neuronas/efectos de los fármacos , Optogenética , Condicionamiento Físico Animal , Proopiomelanocortina/metabolismo , Receptores de Melanocortina/genética , Receptores de Melanocortina/metabolismo , Transducción de Señal , Análisis de la Célula Individual , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/deficiencia , Temperatura
7.
Sci Rep ; 7(1): 7019, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28765602

RESUMEN

Pancreatic ß-cell loss through apoptosis is an important disease mechanism in type 2 diabetes. Apoptosis Repressor with CARD (ARC) is a cell death inhibitor that antagonizes multiple death programs. We previously reported that ARC is abundant in pancreatic ß-cells and modulates survival of these cells in vitro. Herein we assessed the importance of endogenous ARC in maintaining islet structure and function in vivo. While generalized loss of ARC did not result in detectable abnormalities, its absence in ob/ob mice, a model of type 2 diabetes, induced a striking pancreatic phenotype: marked ß-cell death, loss of ß-cell mass, derangements of islet architecture, and impaired glucose-stimulated insulin secretion in vivo. These abnormalities contributed to worsening of hyperglycemia and glucose-intolerance in these mice. Mechanistically, the absence of ARC increased levels of C/EBP homologous protein (CHOP) in wild type isolated islets stimulated with ER stress and in ob/ob isolated islets at baseline. Deletion of CHOP in ob/ob; ARC -/- mice led to reversal of ß-cell death and abnormalities in islet architecture. These data indicate that suppression of CHOP by endogenous levels of ARC is critical for ß-cell viability and maintenance of normal islet structure in this model of type 2 diabetes.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Diabetes Mellitus Tipo 2/patología , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/patología , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción CHOP/metabolismo , Animales , Supervivencia Celular , Modelos Animales de Enfermedad , Secreción de Insulina , Ratones , Ratones Noqueados
8.
Genes Dev ; 29(9): 934-47, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25934505

RESUMEN

MAF1 is a global repressor of RNA polymerase III transcription that regulates the expression of highly abundant noncoding RNAs in response to nutrient availability and cellular stress. Thus, MAF1 function is thought to be important for metabolic economy. Here we show that a whole-body knockout of Maf1 in mice confers resistance to diet-induced obesity and nonalcoholic fatty liver disease by reducing food intake and increasing metabolic inefficiency. Energy expenditure in Maf1(-/-) mice is increased by several mechanisms. Precursor tRNA synthesis was increased in multiple tissues without significant effects on mature tRNA levels, implying increased turnover in a futile tRNA cycle. Elevated futile cycling of hepatic lipids was also observed. Metabolite profiling of the liver and skeletal muscle revealed elevated levels of many amino acids and spermidine, which links the induction of autophagy in Maf1(-/-) mice with their extended life span. The increase in spermidine was accompanied by reduced levels of nicotinamide N-methyltransferase, which promotes polyamine synthesis, enables nicotinamide salvage to regenerate NAD(+), and is associated with obesity resistance. Consistent with this, NAD(+) levels were increased in muscle. The importance of MAF1 for metabolic economy reveals the potential for MAF1 modulators to protect against obesity and its harmful consequences.


Asunto(s)
Proteínas Represoras/genética , Animales , Autofagia/genética , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Metabolismo de los Lípidos/genética , Longevidad/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/genética , ARN de Transferencia/metabolismo , Espermidina/metabolismo
9.
Endocrinology ; 155(11): 4157-67, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25137027

RESUMEN

Although the antidiabetic effects of leptin require intact neuronal melanocortin signaling in rodents with uncontrolled diabetes (uDM), increased melanocortin signaling is not sufficient to mimic leptin's glucose-lowering effects. The current studies were undertaken to clarify the role of melanocortin signaling in leptin's ability to correct metabolic and neuroendocrine disturbances associated with uDM. To accomplish this, bilateral cannulae were implanted in the lateral ventricle of rats with streptozotocin-induced diabetes, and leptin was coinfused with varying doses of the melanocortin 3/4 receptor (MC3/4R) antagonist, SHU9119. An additional cohort of streptozotocin-induced diabetes rats received intracerebroventricular administration of either the MC3/4R agonist, melanotan-II, or its vehicle. Consistent with previous findings, leptin's glucose-lowering effects were blocked by intracerebroventricular SHU9119. In contrast, leptin-mediated suppression of hyperglucagonemia involves both melanocortin dependent and independent mechanisms, and the degree of glucagon inhibition was associated with reduced plasma ketone body levels. Increased central nervous system melanocortin signaling alone fails to mimic leptin's ability to correct any of the metabolic or neuroendocrine disturbances associated with uDM. Moreover, the inability of increased melanocortin signaling to lower diabetic hyperglycemia does not appear to be secondary to release of the endogenous MC3/4R inverse agonist, Agouti-related peptide (AgRP), because AgRP knockout mice did not show increased susceptibility to the antidiabetic effects of increased MC3/4R signaling. Overall, these data suggest that 1) AgRP is not a major driver of diabetic hyperglycemia, 2) mechanisms independent of melanocortin signaling contribute to leptin's antidiabetic effects, and 3) melanocortin receptor blockade dissociates leptin's glucose-lowering effect from its action on other features of uDM, including reversal of hyperglucagonemia and ketosis, suggesting that brain control of ketosis, but not blood glucose levels, is glucagon dependent.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Leptina/uso terapéutico , Melanocortinas/metabolismo , Receptores de Melanocortina/fisiología , Animales , Diabetes Mellitus Experimental/metabolismo , Masculino , Hormonas Estimuladoras de los Melanocitos/farmacología , Ratones , Ratones Noqueados , Sistemas Neurosecretores/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de Melanocortina/antagonistas & inhibidores , Transducción de Señal/fisiología
10.
PLoS One ; 9(2): e89199, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586592

RESUMEN

Mice treated with the antidepressant trans-2-phenylcyclopropylamine (2-PCPA) were protected against diet-induced-obesity, and adiposity was reversed in pre-established diet-induced obese mice. Contrary to a recent report that inhibition of lysine-specific demethylase-1 by 2-PCPA results in increased energy expenditure, long-term 2-PCPA treatment had no such effect but its protection against obesity was associated with increased spontaneous locomotor activity, Moreover, pair feeding to assure equal caloric intake in wild type mice as well as in genetic hyperphagic mice (ob/ob) also resulted in weight reduction in 2-PCPA treated mice that correlated with increased activity but no change in energy expenditure. Similarly, short-term intraperitoneal injections of 2-PCPA did not affect food intake but caused a substantial increase in locomotor activity in the light cycle that correlated with increased energy expenditure, whereas activity and energy expenditure were unchanged in the dark cycle. Lastly, 2-PCPA was also effective in reducing obesity in genetic UCP1 null mice. These data suggest that 2-PCPA can reduce obesity by decreasing food intake in the long term while increasing activity in the short-term. However, the protective and weight loss effects of 2-PCPA are independent of UCP1-regulated thermogenesis or basal energy expenditure.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos/efectos de los fármacos , Obesidad/prevención & control , Tranilcipromina/uso terapéutico , Animales , Composición Corporal/efectos de los fármacos , Ingestión de Energía/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones Obesos , Obesidad/tratamiento farmacológico , Obesidad/etiología , Tranilcipromina/farmacología
11.
Am J Physiol Endocrinol Metab ; 305(12): E1512-20, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24169048

RESUMEN

Deficient leptin signaling causes infertility via reduced activity of GnRH neurons, causing a hypogonadal state in both rodents and humans. Because GnRH neurons do not express leptin receptors, leptin's effect on GnRH neurons must be indirect. Neurons within the hypothalamic arcuate nucleus that coexpress AGRP and NPY are considered to be important intermediate neurons involved in leptin regulation of GnRH neurons. Previously, we reported that the absence of AGRP and haploinsufficiency of MC4R in leptin receptor mutant (Lepr(db/db)) females result in restoration of fertility and lactation despite the persistence of obesity and insulin resistance. The overarching hypothesis in the present study is that the absence or reduction of leptin's inhibition of AGRP/NPY neurons leads to suppression of GnRH release in cases of leptin signaling deficiency. Since TAC2 (NKB)-TAC3R signaling plays a role in puberty maturation and is modulated by metabolic status, the other aim of this study is to test whether TAC2/NKB neurons in ARC regulated by melanocortinergic signals herein affect leptin's action on puberty and reproduction. Our data showed that AGRP deficiency in Lepr(db/db) females restores normal timing of vaginal opening and estrous cycling, although uterine weight gain and mammary gland development are morphologically delayed. Nonetheless, Agrp(-/-) Lepr(db/db) females are fertile and sustain adequate nutrition of pups with lactation to weaning age. AGRP deficiency results in advanced vaginal opening in wild-type female mice. The postpubertal increase in hypothalamic TAC2 mRNA was not observed in Lepr(db/db) females, whereas AGRP deficiency restored it in Lepr(db/db) females. Additionally, MC4R activation with MTII induced FOS expression in TAC2 neurons, supporting the concept of melanocortinergic regulation of TAC2 neurons. These studies suggest that AGRP imposes an inhibitory effect on puberty and that TAC2 neurons may transmit melanocortinergic inhibition of GnRH neurons.


Asunto(s)
Proteína Relacionada con Agouti/fisiología , Leptina/farmacología , Receptores de Leptina/fisiología , Reproducción , Maduración Sexual , Animales , Composición Corporal/efectos de los fármacos , Composición Corporal/genética , Ciclo Estral/efectos de los fármacos , Ciclo Estral/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Reproducción/efectos de los fármacos , Reproducción/genética , Factores Sexuales , Maduración Sexual/efectos de los fármacos , Maduración Sexual/genética
13.
Diabetes ; 62(8): 2784-95, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23557705

RESUMEN

We previously mapped a locus on BALB/c chromosome 2 associated with protection from leptin-deficiency-induced obesity. Here, we generated the corresponding congenic mouse strain by introgression of a segment of C57BL/6J chromosome 2 to the BALB/c background to confirm the genotype-phenotype associations. We found that the BALB/c alleles decreased fat mass expansion by limiting adipocyte hyperplasia and adipocyte hypertrophy. This was concomitant to an increase in adipocyte triglyceride lipase (ATGL)-mediated triglyceride breakdown and prolongation of ATGL half-life in adipose tissue. In addition, BALB/c alleles on chromosome 2 exerted a cell-autonomous role in restraining the adipogenic potential of preadipocytes. Within a 9.8-Mb critical interval, we identified a nonsynonymous coding single nucleotide polymorphism in the gene coding for the ubiquitin-conjugating enzyme E2L6 (Ube2l6, also known as Ubch8) and showed that the BALB/c allele of Ube2l6 is a hypomorph leading to the lack of UBE2L6 protein expression. Ube2l6 knockdown in 3T3-L1 adipocytes repressed adipogenesis. Thus, altered adipogenic potential caused by Ube2l6 knockdown is likely critically involved in BALB/c obesity resistance by inhibiting adipogenesis and reducing adipocyte numbers. Overall, we have identified a loss-of-function mutation in Ube2l6 that contributes to the chromosome 2 obesity quantitative trait locus.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis/genética , Tejido Adiposo/metabolismo , Obesidad/genética , Enzimas Ubiquitina-Conjugadoras/genética , Células 3T3-L1 , Adipocitos/citología , Tejido Adiposo/citología , Alelos , Animales , Glucemia/metabolismo , Diferenciación Celular , Metabolismo Energético , Ratones , Ratones Endogámicos BALB C , Mutación , Obesidad/metabolismo , Sitios de Carácter Cuantitativo , Enzimas Ubiquitina-Conjugadoras/metabolismo
14.
Diabetes ; 62(1): 183-93, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22933109

RESUMEN

Type 2 diabetes involves insulin resistance and ß-cell failure leading to inadequate insulin secretion. An important component of ß-cell failure is cell loss by apoptosis. Apoptosis repressor with caspase recruitment domain (ARC) is an inhibitor of apoptosis that is expressed in cardiac and skeletal myocytes and neurons. ARC possesses the unusual property of antagonizing both the extrinsic (death receptor) and intrinsic (mitochondria/endoplasmic reticulum [ER]) cell death pathways. Here we report that ARC protein is abundant in cells of the endocrine pancreas, including >99.5% of mouse and 73% of human ß-cells. Using genetic gain- and loss-of-function approaches, our data demonstrate that ARC inhibits ß-cell apoptosis elicited by multiple inducers of cell death, including ER stressors tunicamycin, thapsigargin, and physiological concentrations of palmitate. Unexpectedly, ARC diminishes the ER stress response, acting distal to protein kinase RNA-like ER kinase (PERK) and inositol-requiring protein 1α, to suppress C/EBP homologous protein (CHOP) induction. Depletion of ARC in isolated islets augments palmitate-induced apoptosis, which is dramatically rescued by deletion of CHOP. These data demonstrate that ARC is a previously unrecognized inhibitor of apoptosis in ß-cells and that its protective effects are mediated through suppression of the ER stress response pathway.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Estrés del Retículo Endoplásmico , Células Secretoras de Insulina/fisiología , Proteínas Musculares/fisiología , Animales , Apoptosis , Calcio/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Humanos , Ratones , Factor de Transcripción CHOP/fisiología
15.
Int J Neuropsychopharmacol ; 16(4): 857-67, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22932068

RESUMEN

Previous studies have demonstrated that leptin and its receptors (LepRb) in the central nervous system play an important role in regulating depression- and anxiety-related behaviours. However, the physiological functions of LepRb in specific brain regions for mediating different emotional behaviours remain to be defined. In this study, we examined the behavioural effects of LepRb ablation in the adult hippocampus using a series of behavioural paradigms for assessing depression- and anxiety-related behaviours. Targeted deletion of LepRb was achieved using the Cre/loxP site-specific recombination system through bilateral stereotaxic delivery of an adeno-associated virus expressing Cre-recombinase (AAV-Cre) into the dentate gyrus of adult mice homozygous for a floxed leptin receptor allele. AAV-Cre-mediated deletion of the floxed region of LepRb was detected 2 wk after injection. In accordance with this, leptin-stimulated phosphorylation of Akt was attenuated in the hippocampus of AAV-Cre injected mice. Mice injected with AAV-Cre displayed normal locomotor activity and anxiety-like behaviour, as determined in the elevated plus-maze, light-dark box and open field tests, but showed increased depression-like behaviours in the tail suspension, saccharin preference and learned helplessness tests. Taken together, these data suggest that deletion of LepRb in the adult hippocampus is sufficient to induce depression-like behaviours. Our results support the view that leptin signalling in the hippocampus may be essential for positive mood states and active coping to stress.


Asunto(s)
Depresión/metabolismo , Ingestión de Alimentos/fisiología , Ingestión de Alimentos/psicología , Hipocampo/metabolismo , Actividad Motora/fisiología , Receptores de Leptina/deficiencia , Factores de Edad , Animales , Depresión/genética , Depresión/psicología , Ingestión de Alimentos/genética , Hipocampo/fisiopatología , Masculino , Ratones , Ratones Noqueados , Actividad Motora/genética , Receptores de Leptina/genética
16.
Peptides ; 37(1): 6-12, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22800691

RESUMEN

This study used two mouse models with genetic manipulation of the melanocortin system to investigate prolactin regulation. Mice with overexpression of the melanocortin receptor (MC-R) agonist, α-melanocyte-stimulating hormone (Tg-MSH) or deletion of the MC-R antagonist agouti-related protein (AgRP KO) were studied. Male Tg-MSH mice had lower blood prolactin levels at baseline (2.9±0.3 vs. 4.7±0.7ng/ml) and after restraint stress (68±6.5 vs. 117±22ng/ml) vs. WT (p<0.05); however, pituitary prolactin content was not different. Blood prolactin was also decreased in male AgRP KO mice at baseline (4.2±0.5 vs. 7.6±1.3ng/ml) and after stress (60±4.5 vs. 86.1±5.7ng/ml) vs. WT (p<0.001). Pituitary prolactin content was lower in male AgRP KO mice (4.3±0.3 vs. 6.7±0.5µg/pituitary, p<0.001) vs. WT. No differences in blood or pituitary prolactin levels were observed in female AgRP KO mice vs. WT. Hypothalamic dopamine activity was assessed as the potential mechanism responsible for changes in prolactin levels. Hypothalamic tyrosine hydroxylase mRNA was measured in both genetic models vs. WT mice and hypothalamic dopamine and 3,4-dihydroxyphenylacetic acid (DOPAC) content were measured in male AgRP KO and WT mice but neither were significantly different. However, these results do not preclude changes in dopamine activity as dopamine turnover was not directly investigated. This is the first study to show that baseline and stress-induced prolactin release and pituitary prolactin content are reduced in mice with genetic alterations of the melanocortin system and suggests that changes in hypothalamic melanocortin activity may be reflected in measurements of serum prolactin levels.


Asunto(s)
Hipotálamo/metabolismo , Melanocortinas/fisiología , Prolactina/sangre , Ácido 3,4-Dihidroxifenilacético/metabolismo , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Corticosterona/sangre , Dopamina/metabolismo , Antagonistas de Dopamina/farmacología , Femenino , Expresión Génica , Técnicas de Inactivación de Genes , Hipotálamo/enzimología , Masculino , Melanocortinas/metabolismo , Metoclopramida/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hipófisis/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo , alfa-MSH/genética , alfa-MSH/metabolismo
17.
Endocrinology ; 153(5): 2408-19, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22408174

RESUMEN

Leptin and melanocortin signaling control ingestive behavior, energy balance, and substrate utilization, but only leptin signaling defects cause hypothalamic hypogonadism and infertility. Although GnRH neurons do not express leptin receptors, leptin influences GnRH neuron activity via regulation of immediate downstream mediators including the neuropeptides neuropeptide Y and the melanocortin agonist and antagonist, α-MSH, agouti-related peptide, respectively. Here we show that modulation of melanocortin signaling in female db/db mice through ablation of agouti-related peptide, or heterozygosity of melanocortin 4 receptor, restores the timing of pubertal onset, fertility, and lactation. Additionally, melanocortin 4 receptor activation increases action potential firing and induces c-Fos expression in GnRH neurons, providing further evidence that melanocortin signaling influences GnRH neuron activity. These studies thus establish melanocortin signaling as an important component in the leptin-mediated regulation of GnRH neuron activity, initiation of puberty and fertility.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Neuronas/metabolismo , Receptores de Leptina/metabolismo , Maduración Sexual/fisiología , Transducción de Señal/fisiología , Proteína Relacionada con Agouti/genética , Animales , Glucemia/metabolismo , Recuento de Células , Estradiol/sangre , Femenino , Insulina/sangre , Leptina/genética , Ratones , Ratones Noqueados , Ovario/metabolismo , Progesterona/sangre , Receptores de Leptina/genética
18.
Transl Psychiatry ; 2: e83, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22408745

RESUMEN

The glutamatergic system has been implicated in the pathophysiology of depression and the mechanism of action of antidepressants. Leptin, an adipocyte-derived hormone, has antidepressant-like properties. However, the functional role of leptin receptor (Lepr) signaling in glutamatergic neurons remains to be elucidated. In this study, we generated conditional knockout mice in which the long form of Lepr was ablated selectively in glutamatergic neurons located in the forebrain structures, including the hippocampus and prefrontal cortex (Lepr cKO). Lepr cKO mice exhibit normal growth and body weight. Behavioral characterization of Lepr cKO mice reveals depression-like behavioral deficits, including anhedonia, behavioral despair, enhanced learned helplessness and social withdrawal, with no evident signs of anxiety. In addition, loss of Lepr in forebrain glutamatergic neurons facilitates NMDA-induced hippocampal long-term synaptic depression (LTD), whereas conventional LTD or long-term potentiation (LTP) was not affected. The facilitated LTD induction requires activation of the GluN2B subunit as it was completely blocked by a selective GluN2B antagonist. Moreover, Lepr cKO mice are highly sensitive to the antidepressant-like behavioral effects of the GluN2B antagonist but resistant to leptin. These results support important roles for Lepr signaling in glutamatergic neurons in regulating depression-related behaviors and modulating excitatory synaptic strength, suggesting a possible association between synaptic depression and behavioral manifestations of depression.


Asunto(s)
Depresión/fisiopatología , Glutamina/fisiología , Leptina/fisiología , Depresión Sináptica a Largo Plazo/fisiología , Prosencéfalo/fisiopatología , Receptores de Leptina/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Animales , Nivel de Alerta/fisiología , Corteza Cerebral/fisiopatología , Corticosterona/sangre , Dominación-Subordinación , Conducta Exploratoria/fisiología , Desamparo Adquirido , Hipocampo/fisiopatología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Hipotálamo/fisiopatología , Insulina/sangre , Leptina/genética , Ratones , Ratones Noqueados , Motivación/fisiología , Actividad Motora/fisiología , Neuronas/fisiología , Orientación/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Leptina/genética , Receptores de N-Metil-D-Aspartato/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Medio Social , Factores de Transcripción/genética , Factores de Transcripción/fisiología
19.
J Appl Physiol (1985) ; 112(8): 1410-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22323652

RESUMEN

Leptin, a polypeptide hormone produced mainly by adipocytes, has diverse effects in both the brain and peripheral organs, including suppression of feeding. Other than mediating leptin transport across the blood-brain barrier, the role of the endothelial leptin receptor remains unclear. We recently generated a mutant mouse strain lacking endothelial leptin receptor signaling, and showed that there is an increased uptake of leptin by brain parenchyma after its delivery by in situ brain perfusion. Here, we tested the hypothesis that endothelial leptin receptor mutation confers partial resistance to diet-induced obesity. These ELKO mice had similar body weight and percent fat as their wild-type littermates when fed with rodent chow, but blood concentrations of leptin were significantly elevated. In response to a high-fat diet, wild-type mice had a greater gain of body weight and fat than ELKO mice. As shown by metabolic chamber measurement, the ELKO mice had higher oxygen consumption, carbon dioxide production, and heat dissipation, although food intake was similar to that of the wild-type mice and locomotor activity was even reduced. This indicates that the partial resistance to diet-induced obesity was mediated by higher metabolic activity in the ELKO mice. Since neuronal leptin receptor knockout mice show obesity and diabetes, the results suggest that endothelial leptin signaling shows opposite effects from that of neuronal leptin signaling, with a facilitatory role in diet-induced obesity.


Asunto(s)
Grasas de la Dieta/efectos adversos , Endotelio Vascular/metabolismo , Mutación/genética , Obesidad/etiología , Obesidad/prevención & control , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Adiposidad/efectos de los fármacos , Adiposidad/fisiología , Animales , Peso Corporal/fisiología , Dióxido de Carbono/metabolismo , Ritmo Circadiano/fisiología , Grasas de la Dieta/farmacología , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Masculino , Ratones , Ratones Noqueados , Obesidad/metabolismo , Consumo de Oxígeno/fisiología , Receptores de Leptina/metabolismo , Transducción de Señal/fisiología
20.
J Infect Dis ; 205(5): 830-40, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22293433

RESUMEN

Brown adipose tissue (BAT) and white adipose tissue (WAT) and adipocytes are targets of Trypanosoma cruzi infection. Adipose tissue obtained from CD-1 mice 15 days after infection, an early stage of infection revealed a high parasite load. There was a significant increase in macrophages in infected adipose tissue and a reduction in lipid accumulation, adipocyte size, and fat mass and increased expression of lipolytic enzymes. Infection increased levels of Toll-like receptor (TLR) 4 and TLR9 and in the expression of components of the mitogen-activated protein kinase pathway. Protein and messenger RNA (mRNA) levels of peroxisome proliferator-activated receptor γ were increased in WAT, whereas protein and mRNA levels of adiponectin were significantly reduced in BAT and WAT. The mRNA levels of cytokines, chemokines, and their receptors were increased. Nuclear Factor Kappa B levels were increased in BAT, whereas Iκκ-γ levels increased in WAT. Adipose tissue is an early target of T. cruzi infection.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Enfermedad de Chagas/parasitología , ARN Mensajero/metabolismo , Transducción de Señal , Trypanosoma cruzi , Adipocitos/parasitología , Adipocitos/patología , Adiponectina/metabolismo , Tejido Adiposo Pardo/parasitología , Tejido Adiposo Pardo/patología , Tejido Adiposo Blanco/parasitología , Tejido Adiposo Blanco/patología , Animales , Enfermedad de Chagas/patología , Quimiocinas/metabolismo , Citocinas , Quinasa I-kappa B/metabolismo , Masculino , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , PPAR gamma/metabolismo , Receptores de Quimiocina/metabolismo , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 9/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA