Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Cell Rep ; 18(1): 185-197, 2017 01 03.
Article in English | MEDLINE | ID: mdl-28052248

ABSTRACT

Tonotopy is a hallmark of auditory pathways and provides the basis for sound discrimination. Little is known about the involvement of transcription factors in brainstem cochlear neurons orchestrating the tonotopic precision of pre-synaptic input. We found that in the absence of Hoxa2 and Hoxb2 function in Atoh1-derived glutamatergic bushy cells of the anterior ventral cochlear nucleus, broad input topography and sound transmission were largely preserved. However, fine-scale synaptic refinement and sharpening of isofrequency bands of cochlear neuron activation upon pure tone stimulation were impaired in Hox2 mutants, resulting in defective sound-frequency discrimination in behavioral tests. These results establish a role for Hox factors in tonotopic refinement of connectivity and in ensuring the precision of sound transmission in the mammalian auditory circuit.


Subject(s)
Auditory Pathways/physiology , Auditory Perception/physiology , Brain Stem/physiology , Homeodomain Proteins/genetics , Transcription Factors/genetics , Animals , Animals, Newborn , Audiometry, Pure-Tone , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Adhesion , Cochlear Nucleus/physiology , Conditioning, Psychological , Fear , Gene Expression Profiling , Glutamates/metabolism , Homeodomain Proteins/metabolism , Mice , Mice, Mutant Strains , Mutation/genetics , Neurons/metabolism , Organogenesis/genetics , Synapses/metabolism , Synapses/physiology , Synaptic Transmission/physiology , Transcription Factors/metabolism
2.
J Neurosci ; 33(38): 15306-17, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-24048859

ABSTRACT

Neuropeptide Y (NPY) neurons in both the arcuate nucleus of the hypothalamus (ARH) and the dorsomedial hypothalamus (DMH) have been implicated in food intake and obesity. However, while ARH NPY is highly expressed in the lean animal, DMH NPY mRNA expression is observed only after diet-induced obesity (DIO). Furthermore, while ARH NPY neurons are inhibited by leptin, the effect of this adipokine on DMH NPY neurons is unknown. In this study we show that in contrast to the consistent expression in the ARH, DMH NPY mRNA expression was undetectable until after 10 weeks in mice fed a high-fat diet, and peaked at 20 weeks. Surprisingly, electrophysiological experiments demonstrated that leptin directly depolarized and increased the firing rate of DMH NPY neurons in DIO mice. To further differentiate the regulation of DMH and ARH NPY populations, fasting decreased expression of DMH NPY expression, while it increased ARH NPY expression. However, treatment with a leptin receptor antagonist failed to alter DMH NPY expression, indicating that leptin may not be the critical factor regulating mRNA expression. Importantly, we also demonstrated that DMH NPY neurons coexpress cocaine amphetamine-regulated transcript (CART); however, CART mRNA expression in the DMH peaked earlier in the progression of DIO. This study demonstrates novel and important findings. First, NPY and CART are coexpressed in the same neurons within the DMH, and second, leptin stimulates DMH NPY neurons. These studies suggest that during the progression of DIO, there is an unknown signal that drives the expression of the orexigenic NPY signal within the DMH, and that the chronic hyperleptinemia increases the activity of these NPY/CART neurons.


Subject(s)
Gene Expression Regulation/drug effects , Hypothalamus/drug effects , Leptin/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuropeptide Y/metabolism , Obesity/pathology , Action Potentials/drug effects , Action Potentials/genetics , Analysis of Variance , Animals , Diet/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Hypothalamus/pathology , In Vitro Techniques , Insulin/blood , Leptin/antagonists & inhibitors , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neuropeptide Y/genetics , Obesity/blood , Obesity/etiology , Patch-Clamp Techniques , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Radioimmunoassay , STAT3 Transcription Factor/metabolism , Time Factors
3.
J Pediatr Endocrinol Metab ; 26(5-6): 539-44, 2013.
Article in English | MEDLINE | ID: mdl-23509208

ABSTRACT

AIM: We aimed to study the influence of the fat mass and obesity-associated (FTO) gene on eating behavior in 412 obese Sardinian children and adolescents. Genome-wide association studies (GWAS) have identified several susceptibility loci for obesity. Among these, the polymorphisms in the intron 1 of the FTO gene has been found associated to weight gain and obesity in various populations. METHODS: All obese patients were genotyped for the FTO single nucleotide polimorphysm (SNP) rs9939609. In all subjects we evaluated eating behavior using the Child Eating Behaviour Questionnaire (CEBQ). RESULTS: We found no differences in eating behavior according to the genotype, either in the entire cohort, or when subjects were subdivided into four different age groups. CONCLUSIONS: FTO genotype is associated with body mass index but does not influence eating behavior in a selected cohort of obese children from the isolated genetic population of Sardinia.


Subject(s)
Feeding Behavior/physiology , Obesity/genetics , Proteins/genetics , Adolescent , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Body Mass Index , Child , Child, Preschool , Cohort Studies , Female , Gene Frequency , Genome-Wide Association Study , Genotype , Humans , Introns/genetics , Italy , Male , Obesity/epidemiology , Polymorphism, Single Nucleotide/genetics , Prevalence , Young Adult
4.
Science ; 339(6116): 204-7, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23307742

ABSTRACT

We investigated the role of histone methyltransferase Ezh2 in tangential migration of mouse precerebellar pontine nuclei, the main relay between neocortex and cerebellum. By counteracting the sonic hedgehog pathway, Ezh2 represses Netrin1 in dorsal hindbrain, which allows normal pontine neuron migration. In Ezh2 mutants, ectopic Netrin1 derepression results in abnormal migration and supernumerary nuclei integrating in brain circuitry. Moreover, intrinsic topographic organization of pontine nuclei according to rostrocaudal progenitor origin is maintained throughout migration and correlates with patterned cortical input. Ezh2 maintains spatially restricted Hox expression, which, in turn, regulates differential expression of the repulsive receptor Unc5b in migrating neurons; together, they generate subsets with distinct responsiveness to environmental Netrin1. Thus, Ezh2-dependent epigenetic regulation of intrinsic and extrinsic transcriptional programs controls topographic neuronal guidance and connectivity in the cortico-ponto-cerebellar pathway.


Subject(s)
Cerebellum/embryology , Neural Pathways/embryology , Neurons/physiology , Polycomb Repressive Complex 2/metabolism , Pons/embryology , Animals , Cell Movement , Cerebellum/cytology , Cerebellum/metabolism , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Genes, Homeobox , Homeodomain Proteins/metabolism , Metencephalon/embryology , Mice , Mice, Transgenic , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Netrin Receptors , Netrin-1 , Neural Pathways/physiology , Polycomb Repressive Complex 2/genetics , Pons/cytology , Pons/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
5.
Nat Neurosci ; 16(3): 281-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23354331

ABSTRACT

The timing of puberty is controlled by many genes. The elements coordinating this process have not, however, been identified. Here we show that an epigenetic mechanism of transcriptional repression times the initiation of female puberty in rats. We identify silencers of the Polycomb group (PcG) as principal contributors to this mechanism and show that PcG proteins repress Kiss1, a puberty-activating gene. Hypothalamic expression of two key PcG genes, Eed and Cbx7, decreased and methylation of their promoters increased before puberty. Inhibiting DNA methylation blocked both events and resulted in pubertal failure. The pubertal increase in Kiss1 expression was accompanied by EED loss from the Kiss1 promoter and enrichment of histone H3 modifications associated with gene activation. Preventing the eviction of EED from the Kiss1 promoter disrupted pulsatile gonadotropin-releasing hormone release, delayed puberty and compromised fecundity. Our results identify epigenetic silencing as a mechanism underlying the neuroendocrine control of female puberty.


Subject(s)
Epigenesis, Genetic , Hypothalamus/physiology , Sexual Maturation/physiology , Animals , DNA Methylation , Estradiol/blood , Female , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Histones/genetics , Histones/metabolism , Kisspeptins/genetics , Kisspeptins/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Rats , Rats, Sprague-Dawley
6.
J Comp Neurol ; 521(8): 1891-914, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23172177

ABSTRACT

The dorsomedial hypothalamus (DMH) has long been implicated in feeding behavior and thermogenesis. The DMH contains orexigenic neuropeptide Y (NPY) neurons, but the role of these neurons in the control of energy homeostasis is not well understood. NPY expression in the DMH is low under normal conditions in adult rodents but is significantly increased during chronic hyperphagic conditions such as lactation and diet-induced obesity (DIO). To understand better the role of DMH-NPY neurons, we characterized the efferent projections of DMH-NPY neurons using the anterograde tracer biotinylated dextran amine (BDA) in lactating rats and DIO mice. In both models, BDA- and NPY-colabeled fibers were limited mainly to the hypothalamus, including the paraventricular nucleus of the hypothalamus (PVH), lateral hypothalamus/perifornical area (LH/PFA), and anteroventral periventricular nucleus (AVPV). Specifically in lactating rats, BDA-and NPY-colabeled axonal swellings were in close apposition to cocaine- and amphetamine-regulated transcript (CART)-expressing neurons in the PVH and AVPV. Although the DMH neurons project to the rostral raphe pallidus (rRPa), these projections did not contain NPY immunoreactivity in either the lactating rat or the DIO mouse. Instead, the majority of BDA-labeled fibers in the rRPa were orexin positive. Furthermore, DMH-NPY projections were not observed within the nucleus of the solitary tract (NTS), another brainstem site critical for the regulation of sympathetic outflow. The present data suggest that NPY expression in the DMH during chronic hyperphagic conditions plays important roles in feeding behavior and thermogenesis by modulating neuronal functions within the hypothalamus, but not in the brainstem.


Subject(s)
Efferent Pathways/metabolism , Hyperphagia/pathology , Hypothalamus/cytology , Neurons/metabolism , Neuropeptide Y/metabolism , Obesity/pathology , Age Factors , Animals , Animals, Newborn , Biotin/analogs & derivatives , Chronic Disease , Dextrans , Disease Models, Animal , Efferent Pathways/physiology , Female , Gonadotropin-Releasing Hormone/metabolism , Hypothalamic Hormones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lactic Acid/metabolism , Male , Melanins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Obesity/etiology , Orexins , Peptide Fragments/metabolism , Pituitary Hormones/metabolism , Pregnancy , Rats , Rats, Wistar , Tryptophan Hydroxylase/metabolism
7.
Mol Cell Endocrinol ; 351(2): 184-98, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22209758

ABSTRACT

Mammalian puberty is initiated by an increased pulsatile release of gonadotropin-releasing hormone (GnRH) from specialized neurons located in the hypothalamus. GnRH secretion is controlled by neuronal and glial networks, whose activity appears to be coordinated via transcriptional regulation. One of the transcription factors involved in this process is thought to be the recently described gene Enhanced at Puberty 1 (EAP1), which encodes a protein with dual transcriptional activity. In this study we used gene reporter and chromatin immunoprecipitation (ChIP) assays to examine the hypothesis that EAP1 expression is controlled by transcriptional regulators earlier postulated to serve as central nodes of a gene network involved in the neuroendocrine control of puberty. These regulators include Thyroid Transcription Factor 1 (TTF1), Yin Yang 1 (YY1), and CUX1, in addition to EAP1 itself. While TTF1 has been shown to facilitate the advent of puberty, YY1 (a zinc finger protein component of the Polycomb silencing complex) may play a repressive role. The precise role of CUX1 in this context is not known, but like EAP1, CUX1 can either activate or repress gene transcription. We observed that DNA segments of two different lengths (998 and 2744bp) derived from the 5'-flanking region of the human EAP1 gene display similar transcriptional activity. TTF1 stimulates transcription from both DNA segments with equal potency, whereas YY1, CUX1, and EAP1 itself, behave as transcriptional repressors. All four proteins are recruited in vivo to the EAP1 5'-flanking region. These observations suggest that EAP1 gene expression is under dual transcriptional regulation imposed by a trans-activator (TTF1) and two repressors (YY1 and CUX1) previously postulated to be upstream components of a puberty-controlling gene network. In addition, EAP1 itself appears to control its own expression via a negative auto-feedback loop mechanism. Further studies are needed to determine if the occupancy of the EAP1 promoter by these regulatory factors changes at the time of puberty.


Subject(s)
Gene Regulatory Networks , Genes, Regulator/genetics , Genes, Tumor Suppressor , Neoplasm Proteins/genetics , Animals , Binding Sites , Cell Line , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation , Gonadotropin-Releasing Hormone/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypothalamus/metabolism , Hypothalamus/physiology , Neoplasm Proteins/biosynthesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Puberty/genetics , Rats , Rats, Sprague-Dawley , Regulatory Sequences, Nucleic Acid , Repressor Proteins/genetics , Repressor Proteins/metabolism , Securin , Transcription Factors , Transcription, Genetic , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
8.
Ann Hum Genet ; 75(6): 648-54, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21913894

ABSTRACT

Several studies have reported an association of the intronic single nucleotide polymorphism (SNP) rs9939609 of the fat mass and obesity-associated (FTO) gene with obesity and with a number of obesity-related features. We studied the association of rs9939609 with obesity in 912 obese children and adolescents (426 males and 486 females, mean ± SD age 10.5 ± 3.3 years) and in 543 normal weight subjects. A number of biochemical and clinical parameters was also evaluated in 700 of these patients. In the obese group, mean body mass index standard deviation score (BMI-SDS) was similar between the three genotypes. The A allele was present in 55% of the patients' and in 43% of controls' chromosomes. The distribution of heterozygotes was similar between patients and controls (47%), while the distribution of AA homozygotes was significantly higher in patients (31% vs. 20%). Logistic regression analysis on the genotypes yielded a χ(2) of 35.5 with an odds ratio of 1.6 (CI = 1.3-1.8), P < 1 × 10(-5) . None of the clinical and metabolic parameters tested was associated with the genotype. In conclusion, we have confirmed the strong association between FTO and obesity, and shown that only AA homozygotes are predisposed to develop obesity while TT homozygotes might be protected. Finally, we found no association between rs9939609 and a number of obesity-related abnormalities.


Subject(s)
Obesity/genetics , Proteins/genetics , Adolescent , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Case-Control Studies , Child , Female , Genetic Predisposition to Disease , Genotype , Humans , Italy , Male , Polymorphism, Single Nucleotide
9.
Mol Cell Endocrinol ; 342(1-2): 8-19, 2011 Aug 06.
Article in English | MEDLINE | ID: mdl-21672609

ABSTRACT

Kisspeptin, the product of the KiSS1 gene, has emerged as a key component of the mechanism by which the hypothalamus controls puberty and reproductive development. It does so by stimulating the secretion of gonadotropin releasing hormone (GnRH). Little is known about the transcriptional control of the KiSS1 gene. Here we show that a set of proteins postulated to be upstream components of a hypothalamic network involved in controlling female puberty regulates KiSS1 transcriptional activity. Using RACE-PCR we determined that transcription of KiSS1 mRNA is initiated at a single transcription start site (TSS) located 153-156bp upstream of the ATG translation initiation codon. Promoter assays performed using 293 MSR cells showed that the KiSS1 promoter is activated by TTF1 and CUX1-p200, and repressed by EAP1, YY1, and CUX1-p110. EAP1 and CUX-110 were also repressive in GT1-7 cells. All four TFs are recruited in vivo to the KiSS1 promoter and are expressed in kisspeptin neurons. These results suggest that expression of the KiSS1 gene is regulated by trans-activators and repressors involved in the system-wide control of mammalian puberty.


Subject(s)
Gene Expression Regulation , Kisspeptins/genetics , Transcription, Genetic , Chromatin Immunoprecipitation , Female , HeLa Cells , Humans , Hypothalamus/metabolism , Promoter Regions, Genetic/genetics , Puberty/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trans-Activators/metabolism , Transcription Initiation Site
10.
Brain Res ; 1364: 164-74, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20851111

ABSTRACT

The initiation of mammalian puberty requires a sustained increase in pulsatile release of gonadotrophin releasing hormone (GnRH) from the hypothalamus. This increase is brought about by coordinated changes in transsynaptic and glial-neuronal communication, consisting of an increase in neuronal and glial stimulatory inputs to the GnRH neuronal network and the loss of transsynaptic inhibitory influences. GnRH secretion is stimulated by transsynaptic inputs provided by excitatory amino acids (glutamate) and at least one peptide (kisspeptin), and by glial inputs provided by growth factors and small bioactive molecules. The inhibitory input to GnRH neurons is mostly transsynaptic and provided by GABAergic and opiatergic neurons; however, GABA has also been shown to directly excite GnRH neurons. There are many genes involved in the control of these cellular networks, and hence in the control of the pubertal process as a whole. Our laboratory has proposed the concept that these genes are arranged in overlapping networks internally organized in a hierarchical fashion. According to this concept, the highest level of intra-network control is provided by transcriptional regulators that, by directing expression of key subordinate genes, impose genetic coordination to the neuronal and glial subsets involved in initiating the pubertal process. More recently, we have begun to explore the concept that a more dynamic and encompassing level of integrative coordination is provided by epigenetic mechanisms.


Subject(s)
Puberty/genetics , Sexual Maturation/genetics , Transcription, Genetic/physiology , Animals , Epigenomics , Female , Humans , Neurosecretory Systems/physiology , Puberty/physiology , Sexual Maturation/physiology
11.
Ann Hum Genet ; 74(5): 381-6, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20645959

ABSTRACT

Allelic variants of a single nucleotide polymorphism (SNP), rs7566605, located approximately 10 kb upstream of the INSIG2 gene have been found in association with body weight and with other clinical features related to obesity in some populations but not in others. Our objective was to test the association of this SNP in obese children and adolescents from the genetically isolated population of Sardinia. We tested the association of rs7566605 with body mass index (BMI) and with serum glucose and insulin concentrations and a surrogate measure of insulin resistance (HOMA-IR) in a cohort of 747 Sardinian obese children and adolescents. A case control analysis was performed using 548 ethnically-matched healthy controls. Allelic frequencies of the SNP were similar between patients and controls. Mean glucose and insulin concentration and mean HOMA-IR values were significantly higher in patients carrying the CC genotype than in the CG and GG carriers. In the patients with impaired fasting glucose (IFG) and/or impaired glucose tolerance (IGT), allele C was significantly more frequent than in controls. Although INSIG2 polymorphisms do not consistently associate with BMI, the observation of an association with glucose concentration would support a role for this gene in the metabolic complications of obesity.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Obesity/genetics , Obesity/metabolism , Adolescent , Blood Glucose , Female , Humans , Insulin/blood , Insulin Resistance , Italy/epidemiology , Male , Obesity/complications , Obesity/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL