Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 133(7)2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36787185

RESUMEN

Glucose homeostasis can be improved after bariatric surgery, which alters bile flow and stimulates gut hormone secretion, particularly FGF15/19. FGFR1 expression in AGRP-expressing cells is required for bile acids' ability to improve glucose control. We show that the mouse Agrp gene has 3 promoter/enhancer regions that direct transcription of each of their own AGRP transcripts. One of these Agrp promoters/enhancers, Agrp-B, is regulated by bile acids. We generated an Agrp-B knockin FLP/knockout allele. AGRP-B-expressing cells are found in endocrine cells of the pars tuberalis and coexpress diacylglycerol lipase B - an endocannabinoid biosynthetic enzyme - distinct from pars tuberalis thyrotropes. AGRP-B expression is also found in the folliculostellate cells of the pituitary's anterior lobe. Mice without AGRP-B were protected from glucose intolerance induced by high-fat feeding but not from excess weight gain. Chemogenetic inhibition of AGRP-B cells improved glucose tolerance by enhancing glucose-stimulated insulin secretion. Inhibition of the AGRP-B cells also caused weight loss. The improved glucose tolerance and reduced body weight persisted up to 6 weeks after cessation of the DREADD-mediated inhibition, suggesting the presence of a biological switch for glucose homeostasis that is regulated by long-term stability of food availability.


Asunto(s)
Hipotálamo , Neuronas , Ratones , Animales , Proteína Relacionada con Agouti/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Homeostasis , Glucosa/metabolismo , Ácidos y Sales Biliares/metabolismo , Ingestión de Alimentos
2.
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
3.
Biomed Res Int ; 2018: 1085073, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30581841

RESUMEN

Several previous studies have demonstrated the excellent antioxidant activity of fucoxanthin against oxidative stress which is closely related to the pathogenesis of liver diseases. The present work was to investigate whether fucoxanthin could protect human hepatic L02 cells against hydrogen peroxide- (H2O2-) induced oxidative damage. Its effects on H2O2-induced cell viability, lactate dehydrogenase (LDH) leakage, intracellular reduced glutathione, and reactive oxygen species (ROS) contents, along with mRNA and protein relative levels of the cytoprotective genes including Nrf2, HO-1, and NQO1, were investigated. The results showed that fucoxanthin could upregulate the mRNA and protein levels of the cytoprotective genes and promote the nuclear translocation of Nrf2, which could be inhibited by the PI3K inhibitor of LY294002. Pretreatment of fucoxanthin resulted in decreased LDH leakage and intracellular ROS content but enhanced intracellular reduced glutathione. Interestingly, pretreatment using fucoxanthin protected against the oxidative damage in a nonconcentration-dependent manner, with fucoxanthin of 5 µM demonstrating the optimal effects. The results suggest that fucoxanthin exerts cytoprotective effects against H2O2-induced oxidative damage in L02 cells, which may be through the PI3K-dependent activation of Nrf2 signaling.


Asunto(s)
Citoprotección/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Estrés Oxidativo/efectos de los fármacos , Xantófilas/farmacología , Antioxidantes/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cromonas/farmacología , Glutatión/metabolismo , Hemo-Oxigenasa 1/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Morfolinas/farmacología , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Oxidación-Reducción/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
4.
JCI Insight ; 3(17)2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30185666

RESUMEN

When obesity is caused by consumption of a high-fat diet, the tumor suppressor pRb is phosphoinactivated in the neurons of the mediobasal hypothalamus, a brain area critical for energy-balance regulation. However, the functional relevance of pRb phosphoinactivation in the mediobasal hypothalamus to diet-induced obesity remains unknown. Here, we show that inhibiting pRb phosphorylation in the mediobasal hypothalamus can prevent and treat diet-induced obesity in mice. Expressing an unphosphorylable pRb nonselectively in the mediobasal hypothalamus or conditionally in anorexigenic POMC neurons inhibits diet-induced obesity. Intracerebroventricular delivery of US Food and Drug Administration-approved (FDA-approved) cyclin-dependent kinase 4 (CDK4) inhibitor abemaciclib inhibits pRb phosphorylation in the mediobasal hypothalamus and prevents diet-induced obesity. Oral administration of abemaciclib at doses approved for human use reduces fat mass in diet-induced obese mice by increasing lipid oxidation without significantly reducing lean mass. With analysis of recent literature identifying CDK4 as the most abundantly expressed neuronal CDK in the mediobasal hypothalamus, our work uncovers CDK4 as the major kinase for hypothalamic pRb phosphoinactivation and a highly effective central antiobesity target. As three CDK4/6 inhibitors have recently received FDA approval for life-long breast cancer therapy, our study provides a preclinical basis for their expedient repurposing for obesity management.


Asunto(s)
Aminopiridinas/farmacología , Bencimidazoles/farmacología , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Dieta Alta en Grasa/efectos adversos , Obesidad/metabolismo , Obesidad/prevención & control , Animales , Modelos Animales de Enfermedad , Aprobación de Drogas , Metabolismo Energético , Hipotálamo/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Estados Unidos , United States Food and Drug Administration
5.
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
6.
Elife ; 62017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28532548

RESUMEN

Energy dissipation through interscapular brown adipose tissue (iBAT) thermogenesis is an important contributor to adaptive energy expenditure. However, it remains unresolved how acute and chronic changes in energy availability are detected by the brain to adjust iBAT activity and maintain energy homeostasis. Here, we provide evidence that AGRP inhibitory tone to iBAT represents an energy-sparing circuit that integrates environmental food cues and internal signals of energy availability. We establish a role for the nutrient-sensing mTORC1 signaling pathway within AGRP neurons in the detection of environmental food cues and internal signals of energy availability, and in the bi-directional control of iBAT thermogenesis during nutrient deficiency and excess. Collectively, our findings provide insights into how mTORC1 signaling within AGRP neurons surveys energy availability to engage iBAT thermogenesis, and identify AGRP neurons as a neuronal substrate for the coordination of energy intake and adaptive expenditure under varying physiological and environmental contexts.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Metabolismo Energético , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Neuronas/fisiología , Tejido Adiposo/fisiología , Animales , Ratones , Transducción de Señal , Termogénesis
7.
Mol Med Rep ; 10(5): 2681-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25216231

RESUMEN

Aminopeptidase N (APN) is important in tumour processes. The present study detected the anti­tumour activity of the novel APN inhibitor DH­12a, which is an indoline­2,3­dione derivative. In the present study, Bestatin, a clinical APN inhibitor was used as a positive control. The expression of APN in the ES-2 and 3AO cell lines were assessed using flow cytometry and the drug inhibition constants of DH­12a (Ki=13.15 µM) and Bestatin (Ki=16.57 µM) were assessed using a double reciprocal method of competitive inhibition. The in vitro effects of DH­12a on cell proliferation were assessed using a 3­(4,5­dimethyl­thiazol­2­yl)­2,5­diphenyl tetrazolium bromide assay on human cell lines of ES­2 (IC50=43.8 µM), A549 (inhibition rate=41.5% at 160 µM DH­12a), HL60 (inhibition rate=47.83% at 160 µM DH­12a) and 3AO (IC50=70.2 µM). The inhibition rates were consistently higher than those of Bestatin. The effects of DH­12a on cell migration (inhibition rates in ES­2 cells and 3AO cells were 56.4 and 76.5%, respectively at 15 µM) and invasion (inhibition rates in ES­2 cells and 3AO cells were 75.6 and 66.5%, respectively at 15 µM) were assessed using transwell plates. The in vivo effects of DH­12a on tumour proliferation and lung tumour metastasis were determined using an H22 xenograft mice model, where DH­12a was administered in combination with genotoxic 5­fluorouracil. The anti­tumour activities of DH­12a in vivo were also greater than those of Bestatin. In conclusion, the in vitro effects of DH­12a on tumour proliferation, migration and invasion were consistent with the in vivo effects. In addition, DH­12a exhibited greater anti­tumour properties compared with Bestatin.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Hidroxámicos/farmacología , Indoles/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Animales , Antígenos CD13/antagonistas & inhibidores , Antígenos CD13/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Fluorouracilo/farmacología , Concentración 50 Inhibidora , Leucina/análogos & derivados , Leucina/farmacología , Neoplasias Pulmonares/secundario , Ratones , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
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
9.
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
10.
J Neurosci ; 32(29): 9870-7, 2012 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-22815502

RESUMEN

Thioredoxin interacting protein (TXNIP) has recently been described as a key regulator of energy metabolism through pleiotropic actions that include nutrient sensing in the mediobasal hypothalamus (MBH). However, the role of TXNIP in neurochemically specific hypothalamic subpopulations and the circuits downstream from MBH TXNIP engaged to regulate energy homeostasis remain unexplored. To evaluate the metabolic role of TXNIP activity specifically within arcuate Agrp neurons, we generated Agrp-specific TXNIP gain-of-function and loss-of-function mouse models using Agrp-Ires-cre mice, TXNIP (flox/flox) mice, and a lentivector expressing the human TXNIP isoform conditionally in the presence of Cre recombinase. Overexpression of TXNIP in Agrp neurons predisposed to diet-induced obesity and adipose tissue storage by decreasing energy expenditure and spontaneous locomotion, without affecting food intake. Conversely, Agrp neuronal TXNIP deletion protected against diet-induced obesity and adipose tissue storage by increasing energy expenditure and spontaneous locomotion, also without affecting food intake. TXNIP overexpression in Agrp neurons did not primarily affect glycemic control, whereas deletion of TXNIP in Agrp neurons improved fasting glucose levels and glucose tolerance independently of its effects on body weight and adiposity. Bidirectional manipulation of TXNIP expression induced reciprocal changes in central leptin sensitivity and the neural regulation of lipolysis. Together, these results identify a critical role for TXNIP in Agrp neurons in mediating diet-induced obesity through the regulation of energy expenditure and adipose tissue metabolism, independently of food intake. They also reveal a previously unidentified role for Agrp neurons in the brain-adipose axis.


Asunto(s)
Adiposidad/fisiología , Proteína Relacionada con Agouti/metabolismo , Proteínas Portadoras/metabolismo , Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Leptina/farmacología , Neuronas/metabolismo , Tiorredoxinas/metabolismo , Proteína Relacionada con Agouti/genética , Animales , Glucemia/metabolismo , Proteínas Portadoras/genética , Dieta , Ingestión de Alimentos/fisiología , Homeostasis/fisiología , Hipotálamo/efectos de los fármacos , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Obesidad/genética , Obesidad/metabolismo , Tiorredoxinas/genética
11.
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
12.
J Lipid Res ; 53(5): 964-972, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22383686

RESUMEN

Dissecting the genetics of complex traits such as obesity allows the identification of causal genes for disease. Here, we show that the BALB/c mouse strain carries genetic variants that confer resistance to obesity induced by leptin-deficiency or a high-fat diet (HFD). We set out to identify the physiological and genetic bases underlying this phenotype. When compared with C57BL6/J ob/ob mice (B6), BALB/c ob/ob mice exhibited decreased food intake, increased thermogenic capacity, and improved fat catabolism, each of which can potentially modify obesity. Interestingly, analysis of F1 ob/ob (progeny of B6 ob/+ × BALB/c ob+) mice revealed that obesity resistance in BALB/c ob/ob mice principally relied upon improved fat mobilization. This was mechanistically explained by increased adipose triglyceride lipase (ATGL) content in adipocytes, along with increased lipolysis and fatty acid oxidation. We conducted a genome-wide scan and defined a quantitative trait locus (QTL) on chromosome 2. BALB/c alleles on chromosome 2 not only associated with the obesity resistance phenotype but also supported increased ATGL content in adipose tissue. In summary, our study provides evidence that leptin-independent control of adipocyte lipolysis rates directly modifies the balance of macronutrient handling and is sufficient to regulate fat mass in the absence of alterations in food intake and energy expenditure.-Marcelin, G., S-M. Liu, X. Li, G. J. Schwartz, and S. Chua.


Asunto(s)
Tejido Adiposo/metabolismo , Leptina/deficiencia , Lipasa/metabolismo , Lipólisis/genética , Triglicéridos/metabolismo , Adipocitos/metabolismo , Tejido Adiposo/citología , Animales , Metabolismo Basal/genética , Cromosomas de los Mamíferos/genética , Diabetes Mellitus/genética , Diabetes Mellitus/prevención & control , Dieta Alta en Grasa/efectos adversos , Resistencia a la Enfermedad/genética , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Ácidos Grasos/metabolismo , Femenino , Espacio Intracelular/metabolismo , Masculino , Ratones , Obesidad/genética , Obesidad/prevención & control , Oxidación-Reducción , Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo/genética , Especificidad de la Especie
13.
Zhonghua Fu Chan Ke Za Zhi ; 46(9): 655-7, 2011 Sep.
Artículo en Chino | MEDLINE | ID: mdl-22176988

RESUMEN

OBJECTIVE: To investigate the methods and clinical significance of detecting PLAC4 and COL6A1 gene on fetal chromosome 21 from maternal peripheral blood. METHODS: From Oct. 2008 to Nov. 2009 30 normal pregnancies in Weifang People's Hospital were selected as pregnant group, and 9 non-pregnant women were selected as control group. Quantitative real-time PCR was used to determine transcript levels of the target genes (PLAC4 and COL6A1) in blood samples. Correlation between the expression level and gestational age was analyzed. RESULTS: (1) PLAC4 mRNA was detected in peripheral blood of all pregnant women. Its maximum level was 12.760×10(3) copies/ml, whereas the minimum was 2.105×10(3) copies/ml, and the average value is 6.612×10(3) copies/ml. In control group the PLAC4 mRNA could not be detected. There was statistically significant difference (P < 0.01) between the two groups. (2) COL6A1 mRNA is detected in pregnant group and control group, and the concentration was 6.847×10(3) copies/ml and 7.322×10(3) copies/ml respectively, with no statistically significant difference (P > 0.05). (3) Correlation analysis: there was no relationship between the level of PLAC4, COL6A1 mRNA and the gestational age, the correlation coefficients (r) were 0.29 and 0.31, and the P values were 0.121 and 0.168 respectively. CONCLUSIONS: COL6A1 mRNA can be detected in both pregnant group and control group, so it is not specific for pregnancy. PLAC4 mRNA can be detected only in pregnant women, so it has specificity in pregnancy and can be a discriminative marker gene for prenatal diagnosis of trisomy 21 fetuses.


Asunto(s)
Colágeno Tipo VI/sangre , Síndrome de Down/diagnóstico , Proteínas Gestacionales/sangre , Diagnóstico Prenatal/métodos , ARN Mensajero/sangre , Adulto , Biomarcadores/sangre , Estudios de Casos y Controles , Colágeno Tipo VI/genética , Citogenética , Síndrome de Down/sangre , Femenino , Humanos , Reacción en Cadena de la Polimerasa/métodos , Embarazo , Proteínas Gestacionales/genética , ARN Mensajero/genética
14.
Endocrinology ; 152(3): 883-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21285324

RESUMEN

Neuropeptide Y (NPY) and agouti-related peptide (AGRP) can produce hyperphagia, reduce energy expenditure, and promote triglyceride deposition in adipose depots. As these two neuropeptides are coexpressed within the hypothalamic arcuate nucleus and mediate a major portion of the obesity caused by leptin signaling deficiency, we sought to determine whether the two neuropeptides mediated identical or complementary actions. Because of separate neuropeptide receptors and signal transduction mechanisms, there is a possibility of distinct encoding systems for the feeding and energy expenditure aspects of leptin-regulated metabolism. We have genetically added NPY deficiency and/or AGRP deficiency to LEPR deficiency isolated to AGRP cells. Our results indicate that the obesity of LEPR deficiency in AGRP/NPY neurons can produce obesity with either AGRP or NPY alone with AGRP producing hyperphagia while NPY promotes reduced energy expenditure. The absence of both NPY and AGRP prevents the development of obesity attributable to isolated LEPR deficiency in AGRP/NPY neurons. Operant behavioral testing indicated that there were no alterations in the reward for a food pellet from the AGRP-specific LEPR deficiency.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Metabolismo Energético/fisiología , Hiperfagia/metabolismo , Neuropéptido Y/metabolismo , Receptores de Leptina/metabolismo , Adiposidad/genética , Adiposidad/fisiología , Proteína Relacionada con Agouti/genética , Animales , Composición Corporal , Femenino , Hiperfagia/genética , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuropéptido Y/genética , Receptores de Leptina/genética
15.
Kidney Int ; 78(5): 453-62, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20520596

RESUMEN

Most mouse models of diabetes do not fully reproduce features of human diabetic nephropathy, limiting their utility in inferring mechanisms of human disease. Here we performed detailed phenotypic and genetic characterization of leptin-receptor (Lepr) deficient mice on the FVB/NJ background (FVB(db/db)), an obese model of type II diabetes, to determine their suitability to model human diabetic nephropathy. These mice have sustained hyperglycemia, significant albuminuria and characteristic diabetic renal findings including mesangial sclerosis and nodular glomerulosclerosis after 6 months of age. In contrast, equally obese, hyperglycemic Lepr/Sur1 deficient C57BL/6J (Sur1 has defective insulin secretion) mice have minimal evidence of nephropathy. A genome-wide scan in 165 Lepr deficient backcross progeny derived from FVB/NJ and C57BL/6J identified a major locus influencing nephropathy and albuminuria on chromosome 8B1-C5 (Dbnph1 locus, peak lod score 5.0). This locus was distinct from those contrasting susceptibility to beta cell hypertrophy and HIV-nephropathy between the same parental strains, indicating specificity to diabetic kidney disease. Genome-wide expression profiling showed that high and low risk Dbnph1 genotypes were associated with significant enrichment for oxidative phosphorylation and lipid clearance, respectively; molecular pathways shared with human diabetic nephropathy. Hence, we found that the FVB(db/db) mouse recapitulates many clinical, histopathological and molecular features of human diabetic nephropathy. Identifying underlying susceptibility gene(s) and downstream dysregulated pathways in these mice may provide insight into the disease pathogenesis in humans.


Asunto(s)
Mapeo Cromosómico , Diabetes Mellitus Tipo 2/genética , Nefropatías Diabéticas/genética , Predisposición Genética a la Enfermedad , Animales , Diabetes Mellitus Tipo 2/complicaciones , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Ligamiento Genético , Ratones , Ratones Endogámicos C57BL , Ratones Obesos
16.
Endocrinology ; 149(4): 1773-85, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18162515

RESUMEN

Two known types of leptin-responsive neurons reside within the arcuate nucleus: the agouti gene-related peptide (AgRP)/neuropeptide Y (NPY) neuron and the proopiomelanocortin (POMC) neuron. By deleting the leptin receptor gene (Lepr) specifically in AgRP/NPY and/or POMC neurons of mice, we examined the several and combined contributions of these neurons to leptin action. Body weight and adiposity were increased by Lepr deletion from AgRP and POMC neurons individually, and simultaneous deletion in both neurons (A+P LEPR-KO mice) further increased these measures. Young (periweaning) A+P LEPR-KO mice exhibit hyperphagia and decreased energy expenditure, with increased weight gain, oxidative sparing of triglycerides, and increased fat accumulation. Interestingly, however, many of these abnormalities were attenuated in adult animals, and high doses of leptin partially suppress food intake in the A+P LEPR-KO mice. Although mildly hyperinsulinemic, the A+P LEPR-KO mice displayed normal glucose tolerance and fertility. Thus, AgRP/NPY and POMC neurons each play mandatory roles in aspects of leptin-regulated energy homeostasis, high leptin levels in adult mice mitigate the importance of leptin-responsiveness in these neurons for components of energy balance, suggesting the presence of other leptin-regulated pathways that partially compensate for the lack of leptin action on the POMC and AgRP/NPY neurons.


Asunto(s)
Proteína Relacionada con Agouti/fisiología , Ingestión de Alimentos , Metabolismo Energético , Proopiomelanocortina/fisiología , Receptores de Leptina/fisiología , Animales , Composición Corporal , Fertilidad , Hiperinsulinismo/etiología , Hiperfagia , Lactancia , Masculino , Ratones , Neuropéptido Y/fisiología
17.
Endocrinology ; 147(5): 2287-95, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16484328

RESUMEN

Leptin signaling is a critical component of normal insulin sensitivity. Overt hyperglycemia and type 2 diabetes mellitus can be manifested in states of leptin signaling deficiencies by the additional effects of other genetic factors. We have previously described the contrasting insulin sensitivities and glycemic states of two congenic diabetes (db/db) mouse strains. C57BL/6J db/db mice have mild insulin resistance and achieve euglycemia with mild hyperinsulinemia. FVB db/db mice have severe insulin resistance and are hyperglycemic despite escalating hyperinsulinemia with expanded pancreatic beta-cell mass. Analysis of obese progeny from the two reciprocal backcrosses suggests that genetic modifiers for insulin sensitivity are separable from loci that modulate beta-cell mass. A genome scan of the backcross to FVB suggests that one or more modifier genes are present on chromosome 5. This evidence is supported by the phenotypes of multiple incipient congenic strains wherein the hyperglycemia observed in obese FVB mice is reproduced. With similar degrees of hyperglycemia in obese mice of these strains, the haplotype at chromosome 5 is associated with beta-cell mass and circulating insulin concentrations. Finally, we offer arguments that production of multiple incipient congenic lines is an economical alternative to the production of speed congenic strains.


Asunto(s)
Alelos , Variación Genética , Hiperglucemia/genética , Receptores de Superficie Celular/genética , Animales , Mapeo Cromosómico , Cruzamientos Genéticos , Diabetes Mellitus Experimental/genética , Femenino , Genotipo , Glucosa/metabolismo , Haplotipos , Heterocigoto , Hiperglucemia/metabolismo , Insulina/metabolismo , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Modelos Genéticos , Fenotipo , Receptores de Superficie Celular/fisiología , Receptores de Leptina
18.
J Clin Invest ; 115(12): 3484-93, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16284652

RESUMEN

We have generated mice that carry a neuron-specific leptin receptor (LEPR) transgene whose expression is driven by the rat synapsin I promoter synapsin-LEPR B (SYN-LEPR-B). We have also generated mice that are compound hemizygotes for the transgenes SYN-LEPR-B and neuron-specific enolase-LEPR B (NSE-LEPR-B). We observed a degree of correction in db/db mice that are hemizygous (Syn db/db) and homozygous (Syn/Syn db/db) for the SYN-LEPR-B transgene similar to that previously reported for the NSE-LEPR-B transgene. We also show complete correction of the obesity and related phenotypes of db/db mice that are hemizygous for both NSE-LEPR-B and SYN-LEPR-B transgenes (Nse+Syn db/db). Body composition, insulin sensitivity, and cold tolerance were completely normalized in Nse+Syn db/db mice at 12 weeks of age compared with lean controls. In situ hybridization for LEPR B isoform expression in Nse+Syn db/db mice showed robust expression in the energy homeostasis-relevant regions of the hypothalamus. Expression of 3 neuropeptide genes, agouti-related peptide (Agrp), neuropeptide Y (Npy), and proopiomelanocortin (Pomc), was fully normalized in dual transgenic db/db mice. The 2 transgenes in concert conferred normal fertility to male and female db/db mice. Male mice with partial peripheral deletion of Lepr, induced in the periweaning phase, did not show alterations in body composition or mass. In summary, we show that brain-specific leptin signaling is sufficient to reverse the obesity, diabetes, and infertility of db/db mice.


Asunto(s)
Diabetes Mellitus/terapia , Terapia Genética/métodos , Infertilidad/terapia , Neuronas/metabolismo , Obesidad/terapia , Receptores de Superficie Celular/genética , Proteína Relacionada con Agouti , Alelos , Animales , Glucemia/metabolismo , Composición Corporal , Peso Corporal , Frío , ADN Complementario/metabolismo , Diabetes Mellitus/genética , Femenino , Fertilidad , Regulación de la Expresión Génica , Genotipo , Glucosa/metabolismo , Homeostasis , Homocigoto , Hipotálamo/patología , Hibridación in Situ , Infertilidad/genética , Infertilidad Femenina/terapia , Infertilidad Masculina/terapia , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Ratones Transgénicos , Neuropéptido Y/genética , Obesidad/genética , Péptidos/química , Fenotipo , Fosfopiruvato Hidratasa/genética , Reacción en Cadena de la Polimerasa , Proopiomelanocortina/genética , Regiones Promotoras Genéticas , Isoformas de Proteínas , Proteínas/genética , Ratas , Receptores de Leptina , Transducción de Señal , Sinapsinas/genética , Factores de Tiempo , Distribución Tisular , Transgenes
19.
Am J Physiol Endocrinol Metab ; 289(3): E403-11, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15870101

RESUMEN

Leptin signaling in the brain regulates energy intake and expenditure. To test the degree of functional neuronal leptin signaling required for the maintenance of body composition, fertility, and cold tolerance, transgenic mice expressing Cre in neurons (CaMKIIalpha-Cre) were crossed to mice carrying a floxed leptin receptor (Lepr) allele to generate mice with neuron-specific deletion of Lepr in approximately 50% (C F/F mice) and approximately 75% (C Delta17/F mice) of hypothalamic neurons. Leptin receptor (LEPR)-deficient mice (Delta17/Delta17) with heat-shock-Cre-mediated global Lepr deletion served as obese controls. At 16 wk, male C F/F, C Delta17/F, and Delta17/Delta17 mice were 13.2 (P < 0.05), 45.0, and 55.9% (P < 0.001) heavier, respectively, than lean controls, whereas females showed 31.6, 68.8, and 160.7% increases in body mass (P < 0.001). Significant increases in total fat mass (C F/F: P < 0.01; C Delta17/F and Delta17/Delta17:P < 0.001 vs. sex-matched, lean controls), and serum leptin concentrations (P < 0.001 vs. controls) were present in proportion to Lepr deletion. Male C Delta17/F mice had significant elevations in basal serum insulin concentrations (P < 0.001 vs. controls) and were glucose intolerant, as measured by glucose tolerance test (AUC P < 0.01 vs. controls). In contrast with previous observations in mice null for LEPR signaling, C F/F and C Delta17/F mice were fertile and cold tolerant. These findings support the hypothesis that body weight, adiposity, serum leptin concentrations, and glucose intolerance are proportional to hypothalamic LEPR deficiency. However, fertility and cold tolerance remain intact unless hypothalamic LEPR deficiency is complete.


Asunto(s)
Adaptación Fisiológica/fisiología , Frío , Diabetes Mellitus Experimental/fisiopatología , Fertilidad/fisiología , Receptores de Superficie Celular/genética , Tejido Adiposo Pardo/fisiología , Animales , Arginina Vasopresina/genética , Peso Corporal , ADN Complementario , Diabetes Mellitus Experimental/genética , Ingestión de Alimentos , Femenino , Hipotálamo/citología , Hipotálamo/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neuronas/fisiología , Neuropéptido Y/genética , Obesidad/genética , Obesidad/fisiopatología , Proopiomelanocortina/genética , Receptores de Leptina
20.
Mamm Genome ; 15(9): 677-85, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15389315

RESUMEN

Body weight regulation is mediated through several major signaling pathways, some of which have been delineated by positional cloning of spontaneous genetic mutations in mice. Lepr(db/db) mice are obese due to a defect in the signaling portion of the leptin receptor, which has led to extensive study of this highly conserved system over the past several years. We have created an allelic series at Lepr for the further examination of LEPR signaling phenotypes using both the FLP /frt and CRE /loxP systems. By inserting a frt-PGK-neo-frt sequence in Lepr intron 16, we have generated a conditional gene repair Lepr allele ( Lepr-neo) that elicits morbid obesity, diabetes, and infertility in homozygous mice, recapitulating the obesity syndrome of Lepr(db/db) mice. Thus, in vivo excision of the PGK-neo cassette with a FLP recombinase transgene restores the lean and fertile phenotype to Lepr(flox/flox) mice. In the same construct, we have also inserted loxP sites that flank Lepr coding exon 17, a region that encodes a JAK docking site required for STAT3 signaling. CRE-mediated excision of Lepr coding exon 17 from Lepr with a frameshift in subsequent exons results in a syndrome of obesity, diabetes, and infertility in LeprDelta17/Delta17 mice, which is indistinguishable from Lepr(neo/neo) and Lepr(db/db) mice. We conclude that suppression of Lepr gene expression by PGK-neo is phenotypically equivalent to deletion of the Lepr signaling motifs, and therefore the Lepr(neo/neo) mouse may be used to investigate conditional gene repair of Lepr signaling deficiency.


Asunto(s)
Alelos , ADN Nucleotidiltransferasas/genética , Integrasas/genética , Obesidad/genética , Receptores de Superficie Celular/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Glucemia/metabolismo , Regulación de la Temperatura Corporal/genética , Peso Corporal/genética , Peso Corporal/fisiología , Quimera , Frío , Modelos Animales de Enfermedad , Ingestión de Alimentos/genética , Ingestión de Alimentos/fisiología , Femenino , Insulina/sangre , Masculino , Ratones , Datos de Secuencia Molecular , Obesidad/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Superficie Celular/biosíntesis , Receptores de Leptina , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...