Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cell Mol Biol Lett ; 26(1): 5, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33588752

RESUMEN

BACKGROUND: The synaptic vesicle glycoprotein 2 (SV2) family is essential to the synaptic machinery involved in neurotransmission and vesicle recycling. The isoforms SV2A, SV2B and SV2C are implicated in neurological diseases such as epilepsy, Alzheimer's and Parkinson's disease. Suitable cell systems for studying regulation of these proteins are essential. Here we present gene expression data of SV2A, SV2B and SV2C in two human neuroblastoma cell lines after differentiation. METHODS: Human neuroblastoma cell lines SiMa and IMR-32 were treated for seven days with growth supplements (B-27 and N-2), all-trans-retinoic acid (ATRA) or vasoactive intestinal peptide (VIP) and gene expression levels of SV2 and neuronal targets were analyzed. RESULTS: The two cell lines reacted differently to the treatments, and only one of the three SV2 isoforms was affected at a time. SV2B and choline O-acetyltransferase (CHAT) expression was changed in concert after growth supplement treatment, decreasing in SiMa cells while increasing in IMR-32. ATRA treatment resulted in no detected changes in SV2 expression in either cell line while VIP increased both SV2C and dopamine transporter (DAT) in IMR-32 cells. CONCLUSION: The synergistic expression patterns between SV2B and CHAT as well as between SV2C and DAT mirror the connectivity between these targets found in disease models and knock-out animals, although here no genetic alteration was made. These cell lines and differentiation treatments could possibly be used to study SV2 regulation and function.


Asunto(s)
Diferenciación Celular/genética , Regulación Neoplásica de la Expresión Génica , Glicoproteínas de Membrana/genética , Proteínas del Tejido Nervioso/genética , Neuroblastoma/genética , Neuroblastoma/patología , Sitios de Unión , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Regiones Promotoras Genéticas/genética , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción , Tretinoina/farmacología , Péptido Intestinal Vasoactivo/farmacología
2.
Open Biol ; 7(9)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28878041

RESUMEN

Solute carriers (SLCs) are vital as they are responsible for a major part of the molecular transport over lipid bilayers. At present, there are 430 identified SLCs, of which 28 are called atypical SLCs of major facilitator superfamily (MFS) type. These are MFSD1, 2A, 2B, 3, 4A, 4B, 5, 6, 6 L, 7, 8, 9, 10, 11, 12, 13A, 14A and 14B; SV2A, SV2B and SV2C; SVOP and SVOPL; SPNS1, SPNS2 and SPNS3; and UNC93A and UNC93B1. We studied their fundamental properties, and we also included CLN3, an atypical SLC not yet belonging to any protein family (Pfam) clan, because its involvement in the same neuronal degenerative disorders as MFSD8. With phylogenetic analyses and bioinformatic sequence comparisons, the proteins were divided into 15 families, denoted atypical MFS transporter families (AMTF1-15). Hidden Markov models were used to identify orthologues from human to Drosophila melanogaster and Caenorhabditis elegans Topology predictions revealed 12 transmembrane segments (for all except CLN3), corresponding to the common MFS structure. With single-cell RNA sequencing and in situ proximity ligation assay on brain cells, co-expressions of several atypical SLCs were identified. Finally, the transcription levels of all genes were analysed in the hypothalamic N25/2 cell line after complete amino acid starvation, showing altered expression levels for several atypical SLCs.


Asunto(s)
Evolución Molecular , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/clasificación , Neuronas/metabolismo , Secuencia de Aminoácidos , Animales , Transporte Biológico , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Pollos/genética , Pollos/metabolismo , Secuencia Conservada , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Humanos , Hipotálamo/citología , Hipotálamo/metabolismo , Cadenas de Markov , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Neuronas/citología , Filogenia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia , Análisis de Secuencia de ARN , Homología de Secuencia de Aminoácido , Análisis de la Célula Individual , Transcripción Genética , Pez Cebra/genética , Pez Cebra/metabolismo
3.
PLoS One ; 12(2): e0172917, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28235079

RESUMEN

SLC38A9 is characterized as a lysosomal component of the amino acid sensing Ragulator-RAG GTPase complex, controlling the mechanistic target of rapamycin complex 1 (mTORC1). Here, immunohistochemistry was used to map SLC38A9 in mouse brain and staining was detected throughout the brain, in cortex, hypothalamus, thalamus, hippocampus, brainstem and cerebellum. More specifically, immunostaining was found in areas known to be involved in amino acid sensing and signaling pathways e.g. piriform cortex and hypothalamus. SLC38A9 immunoreactivity co-localized with both GABAergic and glutamatergic neurons, but not with astrocytes. SLC38A9 play a key role in the mTORC1 pathway, and therefore we performed in vivo starvation and high-fat diet studies, to measure gene expression alterations in specific brain tissues and in larger brain regions. Following starvation, Slc38a9 was upregulated in brainstem and cortex, and in anterior parts of the brain (Bregma 3.2 to -2.1mm). After high-fat diet, Slc38a9 was specifically upregulated in hypothalamus, while overall downregulation was noticed throughout the brain (Bregma 3.2 to -8.6mm).


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Hipotálamo/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Animales , Encéfalo/metabolismo , Dieta Alta en Grasa , Expresión Génica , Masculino , Ratones Endogámicos C57BL , Inanición/metabolismo , Regulación hacia Arriba
4.
Mol Endocrinol ; 29(9): 1303-19, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26168034

RESUMEN

Several genome-wide association studies have linked the Nudix hydrolase family member nucleoside diphosphate-linked moiety X motif 3 (NUDT3) to obesity. However, the manner of NUDT3 involvement in obesity is unknown, and NUDT3 expression, regulation, and signaling in the central nervous system has not been studied. We performed an extensive expression analysis in mice, as well as knocked down the Drosophila NUDT3 homolog Aps in the nervous system, to determine its effect on metabolism. Detailed in situ hybridization studies in the mouse brain revealed abundant Nudt3 mRNA and protein expression throughout the brain, including reward- and feeding-related regions of the hypothalamus and amygdala, whereas Nudt3 mRNA expression was significantly up-regulated in the hypothalamus and brainstem of food-deprived mice. Knocking down Aps in the Drosophila central nervous system, or a subset of median neurosecretory cells, known as the insulin-producing cells (IPCs), induces hyperinsulinemia-like phenotypes, including a decrease in circulating trehalose levels as well as significantly decreasing all carbohydrate levels under starvation conditions. Moreover, lowering Aps IPC expression leads to a decreased ability to recruit these lipids during starvation. Also, loss of neuronal Aps expression caused a starvation susceptibility phenotype while inducing hyperphagia. Finally, the loss of IPC Aps lowered the expression of Akh, Ilp6, and Ilp3, genes known to be inhibited by insulin signaling. These results point toward a role for this gene in the regulation of insulin signaling, which could explain the robust association with obesity in humans.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Insulina/metabolismo , Obesidad/genética , Pirofosfatasas/genética , Inanición/genética , Ácido Anhídrido Hidrolasas/biosíntesis , Ácido Anhídrido Hidrolasas/metabolismo , Amígdala del Cerebelo/metabolismo , Animales , Línea Celular Tumoral , Drosophila , Proteínas de Drosophila/biosíntesis , Técnicas de Silenciamiento del Gen , Células HCT116 , Células HeLa , Humanos , Hiperinsulinismo/genética , Hiperfagia/genética , Hipotálamo/metabolismo , Hormonas de Insectos/biosíntesis , Insulina/genética , Células Secretoras de Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Células MCF-7 , Masculino , Ratones , Ratones Endogámicos C57BL , Oligopéptidos/biosíntesis , Ácido Pirrolidona Carboxílico/análogos & derivados , ARN Mensajero/biosíntesis , Transducción de Señal/genética , Somatomedinas/biosíntesis , Trehalosa/sangre
5.
PLoS One ; 10(6): e0122061, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26047506

RESUMEN

G protein-coupled receptors (GPCRs) are a class of integral membrane proteins mediating physiological functions fundamental for survival, including energy homeostasis. A few years ago, an amino acid sequence of a novel GPCR gene was identified and named GPR178. In this study, we provide new insights regarding the biological significance of Gpr178 protein, investigating its evolutionary history and tissue distribution as well as examining the relationship between its expression level and feeding status. Our phylogenetic analysis indicated that GPR178 is highly conserved among all animal species investigated, and that GPR178 is not a member of a protein family. Real-time PCR and in situ hybridization revealed wide expression of Gpr178 mRNA in both the brain and periphery, with high expression density in the hypothalamus and brainstem, areas involved in the regulation of food intake. Hence, changes in receptor expression were assessed following several feeding paradigms including starvation and overfeeding. Short-term starvation (12-48h) or food restriction resulted in upregulation of Gpr178 mRNA expression in the brainstem, hypothalamus and prefrontal cortex. Conversely, short-term (48h) exposure to sucrose or Intralipid solutions downregulated Gpr178 mRNA in the brainstem; long-term exposure (10 days) to a palatable high-fat and high-sugar diet resulted in a downregulation of Gpr178 in the amygdala but not in the hypothalamus. Our results indicate that hypothalamic Gpr178 gene expression is altered during acute exposure to starvation or acute exposure to palatable food. Changes in gene expression following palatable diet consumption suggest a possible involvement of Gpr178 in the complex mechanisms of feeding reward.


Asunto(s)
Ingestión de Alimentos , Receptores Acoplados a Proteínas G/metabolismo , Amígdala del Cerebelo/metabolismo , Animales , Evolución Biológica , Encéfalo/metabolismo , Hipotálamo/metabolismo , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Filogenia , Corteza Prefrontal/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/clasificación , Receptores Acoplados a Proteínas G/genética
6.
Gene ; 553(1): 1-6, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25275856

RESUMEN

The Rhodopsin family is a class of integral membrane proteins belonging to G protein-coupled receptors (GPCRs). To date, several orphan GPCRs are still uncharacterized and in this study we present an anatomical characterization of the GPR162 protein and an attempt to describe its functional role. Our results show that GPR162 is widely expressed in GABAergic as well as other neurons within the mouse hippocampus, whereas extensive expression is observed in areas related to energy homeostasis and hedonic feeding such as hypothalamus, amygdala and ventral tegmental area, regions known to be involved in the regulation of palatable food consumption.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Sistema Nervioso Central/metabolismo , Hipotálamo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Área Tegmental Ventral/metabolismo , Amígdala del Cerebelo/fisiología , Animales , Western Blotting , Línea Celular , Conducta Alimentaria , Hipotálamo/fisiología , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño/genética , Receptores Acoplados a Proteínas G/genética , Área Tegmental Ventral/fisiología
7.
PLoS Genet ; 10(9): e1004499, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25187989

RESUMEN

In all animals managing the size of individual meals and frequency of feeding is crucial for metabolic homeostasis. In the current study we demonstrate that the noradrenalin analogue octopamine and the cholecystokinin (CCK) homologue Drosulfakinin (Dsk) function downstream of TfAP-2 and Tiwaz (Twz) to control the number of meals in adult flies. Loss of TfAP-2 or Twz in octopaminergic neurons increased the size of individual meals, while overexpression of TfAP-2 significantly decreased meal size and increased feeding frequency. Of note, our study reveals that TfAP-2 and Twz regulate octopamine signaling to initiate feeding; then octopamine, in a negative feedback loop, induces expression of Dsk to inhibit consummatory behavior. Intriguingly, we found that the mouse TfAP-2 and Twz homologues, AP-2ß and Kctd15, co-localize in areas of the brain known to regulate feeding behavior and reward, and a proximity ligation assay (PLA) demonstrated that AP-2ß and Kctd15 interact directly in a mouse hypothalamus-derived cell line. Finally, we show that in this mouse hypothalamic cell line AP-2ß and Kctd15 directly interact with Ube2i, a mouse sumoylation enzyme, and that AP-2ß may itself be sumoylated. Our study reveals how two obesity-linked homologues regulate metabolic homeostasis by modulating consummatory behavior.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiología , Conducta Alimentaria/fisiología , Comidas/fisiología , Obesidad/metabolismo , Obesidad/fisiopatología , Animales , Línea Celular , Retroalimentación , Homeostasis/fisiología , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Octopamina/metabolismo , Canales de Potasio/metabolismo , Factor de Transcripción AP-2/metabolismo
8.
Brain Res ; 1557: 12-25, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24530433

RESUMEN

PAT4, the fourth member of the SLC36/proton dependent amino acid transporter (PAT) family, is a high-affinity, low capacity electroneutral transporter of neutral amino acids like proline and tryptophan. It has also been associated with the function of mTORC1, a complex in the mammalian target of rapamycin (mTOR) pathway. We performed in situ hybridization and immunohistological analysis to determine the expression profile of PAT4, as well as an RT-PCR study on tissue from mice exposed to leucine. We performed a phylogenetic analysis to determine the evolutionary origin of PAT4. The in situ hybridization and the immunohistochemistry on mouse brain sections and hypothalamic cells showed abundant PAT4 expression in the mouse brain intracellularly in both inhibitory and excitatory neurons, partially co-localizing with lysosomal markers and epithelial cells lining the ventricles. Its location in epithelial cells around the ventricles indicates a transport of substrates across the blood brain barrier. Phylogenetic analysis showed that PAT4 belongs to an evolutionary old family most likely predating animals, and PAT4 is the oldest member of that family.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Encéfalo/metabolismo , Plexo Coroideo/metabolismo , Células Epiteliales/metabolismo , Neuronas/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Animales , Western Blotting , Encéfalo/irrigación sanguínea , Membrana Celular/metabolismo , Expresión Génica , Hipocampo/irrigación sanguínea , Hipocampo/metabolismo , Hipotálamo/irrigación sanguínea , Hipotálamo/metabolismo , Inmunohistoquímica , Hibridación in Situ , Leucina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Filogenia , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
PLoS Genet ; 8(3): e1002568, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22438821

RESUMEN

Neurobeachin (Nbea) regulates neuronal membrane protein trafficking and is required for the development and functioning of central and neuromuscular synapses. In homozygous knockout (KO) mice, Nbea deficiency causes perinatal death. Here, we report that heterozygous KO mice haploinsufficient for Nbea have higher body weight due to increased adipose tissue mass. In several feeding paradigms, heterozygous KO mice consumed more food than wild-type (WT) controls, and this consumption was primarily driven by calories rather than palatability. Expression analysis of feeding-related genes in the hypothalamus and brainstem with real-time PCR showed differential expression of a subset of neuropeptide or neuropeptide receptor mRNAs between WT and Nbea+/- mice in the sated state and in response to food deprivation, but not to feeding reward. In humans, we identified two intronic NBEA single-nucleotide polymorphisms (SNPs) that are significantly associated with body-mass index (BMI) in adult and juvenile cohorts. Overall, data obtained in mice and humans suggest that variation of Nbea abundance or activity critically affects body weight, presumably by influencing the activity of feeding-related neural circuits. Our study emphasizes the importance of neural mechanisms in body weight control and points out NBEA as a potential risk gene in human obesity.


Asunto(s)
Índice de Masa Corporal , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Conducta Alimentaria , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Obesidad/genética , Tejido Adiposo/metabolismo , Adolescente , Animales , Tronco Encefálico/metabolismo , Niño , Privación de Alimentos , Regulación de la Expresión Génica/genética , Estudios de Asociación Genética , Humanos , Hipotálamo/metabolismo , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple
10.
BMC Neurosci ; 12: 117, 2011 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-22087873

RESUMEN

BACKGROUND: The Fat mass and obesity gene (FTO) has been identified through genome wide association studies as an important genetic factor contributing to a higher body mass index (BMI). However, the molecular context in which this effect is mediated has yet to be determined. We investigated the potential molecular network for FTO by analyzing co-expression and protein-protein interaction databases, Coxpresdb and IntAct, as well as the functional coupling predicting multi-source database, FunCoup. Hypothalamic expression of FTO-linked genes defined with this bioinformatics approach was subsequently studied using quantitative real time-PCR in mouse feeding models known to affect FTO expression. RESULTS: We identified several candidate genes for functional coupling to FTO through database studies and selected nine for further study in animal models. We observed hypothalamic expression of Profilin 2 (Pfn2), cAMP-dependent protein kinase catalytic subunit beta (Prkacb), Brain derived neurotrophic factor (Bdnf), neurotrophic tyrosine kinase, receptor, type 2 (Ntrk2), Signal transducer and activator of transcription 3 (Stat3), and Btbd12 to be co-regulated in concert with Fto. Pfn2 and Prkacb have previously not been linked to feeding regulation. CONCLUSIONS: Gene expression studies validate several candidates generated through database studies of possible FTO-interactors. We speculate about a wider functional role for FTO in the context of current and recent findings, such as in extracellular ligand-induced neuronal plasticity via NTRK2/BDNF, possibly via interaction with the transcription factor CCAAT/enhancer binding protein ß (C/EBPß).


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Hipotálamo/metabolismo , Glicoproteínas de Membrana/genética , Oxigenasas de Función Mixta/genética , Obesidad/genética , Oxo-Ácido-Liasas/genética , Proteínas Tirosina Quinasas/genética , Transducción de Señal/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Animales , Biología Computacional/métodos , Estudio de Asociación del Genoma Completo/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo
11.
FEBS J ; 278(24): 4881-94, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21981325

RESUMEN

The Rhodopsin family of G protein coupled receptors (GPCRs) includes the phylogenetic α-group consisting of about 100 human members. The α-group is the only group of GPCRs that has many receptors for biogenic amines which are major drug targets. Several members of this group are orphan receptors and their functions are elusive. In this study we present a detailed phylogenetic and anatomical characterization of the Gpr153 receptor and also attempt to study its functional role. We identified the homologue of Gpr153 in the elephant shark genome and phylogenetic and synteny analyses revealed that Gpr162 and Gpr153 share a common ancestor that split most likely through a duplication event before the divergence of the tetrapods and the teleost lineage. A quantitative real-time PCR study reveals widespread expression of Gpr153 in the central nervous system and all the peripheral tissues investigated. Detailed in situ hybridization on mouse brain showed specifically high expression in the thalamus, cerebellum and the arcuate nucleus. The antisense oligodeoxynucleotide knockdown of Gpr153 caused a slight reduction in food intake and the elevated plus maze test showed significant reduction in the percentage of time spent in the centre square, which points towards a probable role in decision making. This report provides the first detailed characterization of the evolution, expression and primary functional properties of the Gpr153 gene.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Cerebelo/metabolismo , Receptores Acoplados a Proteínas G/genética , Tálamo/metabolismo , Secuencia de Aminoácidos , Animales , Conducta Animal/efectos de los fármacos , Ingestión de Alimentos , Evolución Molecular , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Datos de Secuencia Molecular , Filogenia , Ratas , Receptores Acoplados a Proteínas G/biosíntesis , Alineación de Secuencia , Tiburones/genética , Sintenía
12.
BMC Neurosci ; 10: 129, 2009 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-19860904

RESUMEN

BACKGROUND: Polymorphism in the FTO gene is strongly associated with obesity, but little is known about the molecular bases of this relationship. We investigated whether hypothalamic FTO is involved in energy-dependent overconsumption of food. We determined FTO mRNA levels in rodent models of short- and long-term intake of palatable fat or sugar, deprivation, diet-induced increase in body weight, baseline preference for fat versus sugar as well as in same-weight animals differing in the inherent propensity to eat calories especially upon availability of diverse diets, using quantitative PCR. FTO gene expression was also studied in organotypic hypothalamic cultures treated with anorexigenic amino acid, leucine. In situ hybridization (ISH) was utilized to study FTO signal in reward- and hunger-related sites, colocalization with anorexigenic oxytocin, and c-Fos immunoreactivity in FTO cells at initiation and termination of a meal. RESULTS: Deprivation upregulated FTO mRNA, while leucine downregulated it. Consumption of palatable diets or macronutrient preference did not affect FTO expression. However, the propensity to ingest more energy without an effect on body weight was associated with lower FTO mRNA levels. We found that 4-fold higher number of FTO cells displayed c-Fos at meal termination as compared to initiation in the paraventricular and arcuate nuclei of re-fed mice. Moreover, ISH showed that FTO is present mainly in hunger-related sites and it shows a high degree of colocalization with anorexigenic oxytocin. CONCLUSION: We conclude that FTO mRNA is present mainly in sites related to hunger/satiation control; changes in hypothalamic FTO expression are associated with cues related to energy intake rather than feeding reward. In line with that, neurons involved in feeding termination express FTO. Interestingly, baseline FTO expression appears linked not only with energy intake but also energy metabolism.


Asunto(s)
Ingestión de Energía/fisiología , Conducta Alimentaria/fisiología , Hipotálamo/metabolismo , Oxo-Ácido-Liasas/metabolismo , Recompensa , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Análisis de Varianza , Animales , Peso Corporal , Dieta , Ingestión de Alimentos/fisiología , Emulsiones Grasas Intravenosas/administración & dosificación , Hipotálamo/efectos de los fármacos , Hibridación in Situ , Leucina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Oxigenasas de Función Mixta , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Técnicas de Cultivo de Órganos , Oxo-Ácido-Liasas/genética , Oxitocina/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sacarosa/administración & dosificación
13.
Peptides ; 29(9): 1588-95, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18550224

RESUMEN

Our understanding of the central regulation of food intake and body weight has increased tremendously through implication of a high number of neuropeptides. However, lack of all-embracing studies have made comparison difficult in the past. The objective of this study was to demonstrate the relative importance of the different neuropeptides in terms of involvement in appetite regulatory mechanisms. We quantified expression levels of 21 hypothalamic neuropeptides and circulating levels of leptin, insulin, corticosterone, adrenocorticotropic hormone, ghrelin and adiponectin in rats after acute food deprivation and chronic food restriction using validated quantitative real-time PCR and hormone measurements. Body weight, insulin and leptin were reduced whereas corticosterone was increased by both acute food deprivation and chronic food restriction. Our results confirmed the relative importance in body weight homeostasis of neuropeptide Y and proopiomelanocortin, which were increased and decreased as predicted. The expression of other neuropeptides previously attributed central roles in body weight homeostasis, e.g. melanin-concentrating hormone and orexin, appeared to be less affected by the treatments. Moreover, the expression of dynorphin, galanin-like peptide and neuropeptide B was dramatically reduced after both treatments. This suggests that the latter neuropeptides--although previously known to be involved in body weight homeostasis--may be of unexpected importance in states of negative energy balance.


Asunto(s)
Ingestión de Energía/fisiología , Privación de Alimentos/fisiología , Hormonas/sangre , Hipotálamo/metabolismo , Neuropéptidos/biosíntesis , Animales , Glucemia/metabolismo , Dinorfinas/sangre , Péptido Similar a Galanina/sangre , Masculino , Neuropéptidos/sangre , Ratas , Ratas Sprague-Dawley
14.
Brain Res Mol Brain Res ; 133(1): 37-46, 2005 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-15661363

RESUMEN

Food restriction is associated with a number of endocrine disturbances. We validated the experimental conditions for several house-keeping genes and determined the effects of 12 day 50% food restriction on hypothalamic and pituitary transcription of genes involved in different neuroendocrine systems, using real-time quantitative polymerase chain reaction (PCR). A total of 7 nuclear receptors and 12 neuropeptides and peptide hormones were investigated in the dorsal and ventral hypothalamus and the pituitary gland in rats. In the hypothalamus, food restriction reduced mRNA levels of estrogen receptor alpha (ERalpha), progesterone receptor, glucocorticoid receptor, thyroid hormone receptor alpha and beta, pro-opiomelanocortin (POMC), growth hormone-releasing factor (GHRF), corticotropin-releasing factor (CRF), thyrotropin-releasing factor (TRF), somatostatin, and increased that of neuropeptide Y (NPY). In the pituitary, the treatment reduced growth hormone (GH), luteinizing hormone beta (LHbeta) and thyrotropin beta, but increased ERalpha mRNA levels. The study provides a map of how food restriction affects the regulation of a number of transcripts involved in neuroendocrine control.


Asunto(s)
Privación de Alimentos/fisiología , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Neuropéptidos/metabolismo , Hormonas Peptídicas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Peso Corporal/fisiología , ADN Complementario/biosíntesis , Ingestión de Alimentos/fisiología , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Sistema Hipófiso-Suprarrenal/metabolismo , ARN/aislamiento & purificación , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
15.
Ann N Y Acad Sci ; 994: 74-83, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12851300

RESUMEN

The melanocortin (MC)-4 receptor participates in regulating body weight homeostasis. We demonstrated early that acute blockage of the MC-4 receptor increases food intake and relieves anorexic conditions in rats. Our recent studies show that 4-week chronic blockage of the MC-4 receptor leads to robust increases in food intake and development of obesity, whereas stimulation of the receptor leads to anorexia. Interestingly, the food conversion ratio was clearly increased by MC-4 receptor blockage, whereas it was decreased in agonist-treated rats in a transient manner. Chronic infusion of an agonist caused a transient increase in oxygen consumption. Our studies also show that the MC-4 receptor plays a role in luteinizing hormone and prolactin surges in female rats. The MC-4 receptor has a role in mediating the effects of leptin on these surges. The phylogenetic relation of the MC-4 receptor to other GPCRs in the human genome was determined. The three-dimensional structure of the protein was studied by construction of a high-affinity zinc binding site between the helices, using two histidine residues facing each other. We also cloned the MC-4 receptor from evolutionary important species and showed by chromosomal mapping a conserved synteny between humans and zebrafish. The MC-4 receptor has been remarkably conserved in structure and pharmacology for more than 400 million years, implying that the receptor participated in vital physiological functions early in vertebrate evolution.


Asunto(s)
Ingestión de Alimentos , Receptores de Corticotropina/metabolismo , Animales , Humanos , Hipotálamo/metabolismo , Metales/metabolismo , Filogenia , Receptor de Melanocortina Tipo 4 , Receptores de Corticotropina/química , Receptores de Corticotropina/clasificación , Receptores de Corticotropina/genética , Reproducción/fisiología , alfa-MSH/agonistas , alfa-MSH/metabolismo
16.
Endocrinology ; 143(5): 1950-63, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11956178

RESUMEN

We have recently shown that hydroxysteroid sulfotransferase (HST), the enzyme responsible for the biosynthesis of pregnenolone sulfate (Delta(5)PS) and dehydroepiandrosterone sulfate (DHEAS), is expressed in neurons located in the anterior preoptic area and the dorsal magnocellular nucleus of the frog diencephalon. As these two nuclei are richly innervated by NPY-immunoreactive fibers, we investigated the possible implication of NPY in the control of Delta(5)PS and DHEAS biosynthesis. Double labeling of frog brain sections revealed that 42% of the HST-immunoreactive perikarya in the diencephalon were contacted by NPY-containing fibers. In situ hybridization studies showed that Y(1) and Y(5) receptor mRNAs are expressed in the anterior preoptic area and the dorsal magnocellular nucleus. Pulse-chase experiments with (35)S-labeled 3'-phosphoadenosine 5'-phosphosulfate as a sulfate donor demonstrated that frog NPY (fNPY) inhibited the conversion of [(3)H]Delta(5)P and [(3)H]dehydroepiandrosterone ([(3)H]DHEA) into [(3)H,(35)S]Delta(5)PS and [(3)H,(35)S]DHEAS by diencephalic explants. The inhibitory effect of fNPY on Delta(5)PS and DHEAS formation was mimicked by (pPYY) and [Leu(31),Pro(34)]pNPY, which is an agonist for non-Y(2) receptors in mammals, and was completely suppressed by the Y(1) receptor antagonist BIBP3226. Conversely, the Y(2) receptor agonist pNPY-(13-36) and the Y(5) receptor agonist [D-Trp(32)]pNPY did not significantly modify the biosynthesis of [(3)H,(35)S]Delta(5)PS and [(3)H,(35)S]DHEAS. The present study provides the first evidence for the innervation of neurosteroid-producing neurons by NPY fibers. Our data also demonstrate that NPY, acting via Y(1) receptors, exerts an inhibitory effect on the biosynthesis of sulfated neurosteroids.


Asunto(s)
Hipotálamo/metabolismo , Neuropéptido Y/farmacología , Receptores de Neuropéptido Y/agonistas , Esteroides/antagonistas & inhibidores , Esteroides/biosíntesis , Animales , Southern Blotting , Western Blotting , Cromatografía Líquida de Alta Presión , Sulfato de Deshidroepiandrosterona/metabolismo , Diencéfalo/efectos de los fármacos , Diencéfalo/metabolismo , Técnica del Anticuerpo Fluorescente , Cobayas , Hipotálamo/efectos de los fármacos , Inmunohistoquímica , Hibridación in Situ , Técnicas In Vitro , Indicadores y Reactivos , Masculino , Oligonucleótidos/biosíntesis , Oligonucleótidos/genética , Rana ridibunda , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfatos/metabolismo , Sulfotransferasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA