Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Exp Med ; 220(11)2023 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-37642942

RESUMO

Pervasive neuroinflammation occurs in many neurodegenerative diseases, including Alzheimer's disease (AD). SPI1/PU.1 is a transcription factor located at a genome-wide significant AD-risk locus and its reduced expression is associated with delayed onset of AD. We analyzed single-cell transcriptomic datasets from microglia of human AD patients and found an enrichment of PU.1-binding motifs in the differentially expressed genes. In hippocampal tissues from transgenic mice with neurodegeneration, we found vastly increased genomic PU.1 binding. We then screened for PU.1 inhibitors using a PU.1 reporter cell line and discovered A11, a molecule with anti-inflammatory efficacy and nanomolar potency. A11 regulated genes putatively by recruiting a repressive complex containing MECP2, HDAC1, SIN3A, and DNMT3A to PU.1 motifs, thus representing a novel mechanism and class of molecules. In mouse models of AD, A11 ameliorated neuroinflammation, loss of neuronal integrity, AD pathology, and improved cognitive performance. This study uncovers a novel class of anti-inflammatory molecules with therapeutic potential for neurodegenerative disorders.


Assuntos
Doença de Alzheimer , Doenças Neuroinflamatórias , Animais , Camundongos , Humanos , Oncogenes , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Linhagem Celular , Modelos Animais de Doenças , Camundongos Transgênicos
2.
Cell Syst ; 6(4): 424-443.e7, 2018 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-29655704

RESUMO

Although the value of proteomics has been demonstrated, cost and scale are typically prohibitive, and gene expression profiling remains dominant for characterizing cellular responses to perturbations. However, high-throughput sentinel assays provide an opportunity for proteomics to contribute at a meaningful scale. We present a systematic library resource (90 drugs × 6 cell lines) of proteomic signatures that measure changes in the reduced-representation phosphoproteome (P100) and changes in epigenetic marks on histones (GCP). A majority of these drugs elicited reproducible signatures, but notable cell line- and assay-specific differences were observed. Using the "connectivity" framework, we compared signatures across cell types and integrated data across assays, including a transcriptional assay (L1000). Consistent connectivity among cell types revealed cellular responses that transcended lineage, and consistent connectivity among assays revealed unexpected associations between drugs. We further leveraged the resource against public data to formulate hypotheses for treatment of multiple myeloma and acute lymphocytic leukemia. This resource is publicly available at https://clue.io/proteomics.


Assuntos
Bases de Dados Factuais , Fosfoproteínas/efeitos dos fármacos , Algoritmos , Linhagem Celular , Cromatografia Líquida , Conjuntos de Dados como Assunto , Regulação da Expressão Gênica , Código das Histonas , Humanos , Espectrometria de Massas , Fenômenos Farmacológicos e Toxicológicos , Fosfoproteínas/metabolismo , Proteômica , Transdução de Sinais , Software
3.
PLoS One ; 11(9): e0161969, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27622770

RESUMO

The dismal success rate of clinical trials for Alzheimer's disease (AD) motivates us to develop model systems of AD pathology that have higher predictive validity. The advent of induced pluripotent stem cells (iPSCs) allows us to model pathology and study disease mechanisms directly in human neural cells from healthy individual as well as AD patients. However, two-dimensional culture systems do not recapitulate the complexity of neural tissue, and phenotypes such as extracellular protein aggregation are difficult to observe. We report brain organoids that use pluripotent stem cells derived from AD patients and recapitulate AD-like pathologies such as amyloid aggregation, hyperphosphorylated tau protein, and endosome abnormalities. These pathologies are observed in an age-dependent manner in organoids derived from multiple familial AD (fAD) patients harboring amyloid precursor protein (APP) duplication or presenilin1 (PSEN1) mutation, compared to controls. The incidence of AD pathology was consistent amongst several fAD lines, which carried different mutations. Although these are complex assemblies of neural tissue, they are also highly amenable to experimental manipulation. We find that treatment of patient-derived organoids with ß- and γ-secretase inhibitors significantly reduces amyloid and tau pathology. Moreover, these results show the potential of this model system to greatly increase the translatability of pre-clinical drug discovery in AD.


Assuntos
Doença de Alzheimer/patologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/citologia , Encéfalo/patologia , Linhagem Celular , Endossomos/patologia , Humanos , Organoides/citologia , Organoides/patologia , Fenótipo , Fosforilação , Alicerces Teciduais , Proteínas tau/metabolismo
5.
Mol Cell Neurosci ; 73: 13-31, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26657644

RESUMO

In the last decade, induced pluripotent stem (iPS) cells have revolutionized the utility of human in vitro models of neurological disease. The iPS-derived and differentiated cells allow researchers to study the impact of a distinct cell type in health and disease as well as performing therapeutic drug screens on a human genetic background. In particular, clinical trials for Alzheimer's disease (AD) have been failing. Two of the potential reasons are first, the species gap involved in proceeding from initial discoveries in rodent models to human studies, and second, an unsatisfying patient stratification, meaning subgrouping patients based on the disease severity due to the lack of phenotypic and genetic markers. iPS cells overcome this obstacles and will improve our understanding of disease subtypes in AD. They allow researchers conducting in depth characterization of neural cells from both familial and sporadic AD patients as well as preclinical screens on human cells. In this review, we briefly outline the status quo of iPS cell research in neurological diseases along with the general advantages and pitfalls of these models. We summarize how genome-editing techniques such as CRISPR/Cas9 will allow researchers to reduce the problem of genomic variability inherent to human studies, followed by recent iPS cell studies relevant to AD. We then focus on current techniques for the differentiation of iPS cells into neural cell types that are relevant to AD research. Finally, we discuss how the generation of three-dimensional cell culture systems will be important for understanding AD phenotypes in a complex cellular milieu, and how both two- and three-dimensional iPS cell models can provide platforms for drug discovery and translational studies into the treatment of AD.


Assuntos
Doença de Alzheimer/patologia , Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Diferenciação Celular , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Modelos Biológicos
6.
Nat Neurosci ; 18(7): 1008-16, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26005852

RESUMO

Noncoding variants in the human MIR137 gene locus increase schizophrenia risk with genome-wide significance. However, the functional consequence of these risk alleles is unknown. Here we examined induced human neurons harboring the minor alleles of four disease-associated single nucleotide polymorphisms in MIR137. We observed increased MIR137 levels compared to those in major allele-carrying cells. microRNA-137 gain of function caused downregulation of the presynaptic target genes complexin-1 (Cplx1), Nsf and synaptotagmin-1 (Syt1), leading to impaired vesicle release. In vivo, miR-137 gain of function resulted in changes in synaptic vesicle pool distribution, impaired induction of mossy fiber long-term potentiation and deficits in hippocampus-dependent learning and memory. By sequestering endogenous miR-137, we were able to ameliorate the synaptic phenotypes. Moreover, reinstatement of Syt1 expression partially restored synaptic plasticity, demonstrating the importance of Syt1 as a miR-137 target. Our data provide new insight into the mechanism by which miR-137 dysregulation can impair synaptic plasticity in the hippocampus.


Assuntos
Regulação da Expressão Gênica/genética , MicroRNAs/metabolismo , Fibras Musgosas Hipocampais/metabolismo , Plasticidade Neuronal/genética , Esquizofrenia/genética , Vesículas Sinápticas/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Alelos , Animais , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Fibroblastos , Loci Gênicos , Células HEK293 , Humanos , Aprendizagem/fisiologia , Potenciação de Longa Duração , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Sensíveis a N-Etilmaleimida/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Polimorfismo de Nucleotídeo Único , Sinaptotagmina I/metabolismo
7.
Proc Natl Acad Sci U S A ; 112(23): 7291-6, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-25995364

RESUMO

Repeated stress has been suggested to underlie learning and memory deficits via the basolateral amygdala (BLA) and the hippocampus; however, the functional contribution of BLA inputs to the hippocampus and their molecular repercussions are not well understood. Here we show that repeated stress is accompanied by generation of the Cdk5 (cyclin-dependent kinase 5)-activator p25, up-regulation and phosphorylation of glucocorticoid receptors, increased HDAC2 expression, and reduced expression of memory-related genes in the hippocampus. A combination of optogenetic and pharmacosynthetic approaches shows that BLA activation is both necessary and sufficient for stress-associated molecular changes and memory impairments. Furthermore, we show that this effect relies on direct glutamatergic projections from the BLA to the dorsal hippocampus. Finally, we show that p25 generation is necessary for the stress-induced memory dysfunction. Taken together, our data provide a neural circuit model for stress-induced hippocampal memory deficits through BLA activity-dependent p25 generation.


Assuntos
Complexo Nuclear Basolateral da Amígdala/fisiopatologia , Quinase 5 Dependente de Ciclina/metabolismo , Hipocampo/fisiopatologia , Deficiências da Aprendizagem/fisiopatologia , Transtornos da Memória/fisiopatologia , Animais , Complexo Nuclear Basolateral da Amígdala/efeitos da radiação , Hipocampo/efeitos da radiação , Luz , Camundongos , Estresse Fisiológico
8.
Cell ; 157(2): 486-498, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24725413

RESUMO

Cyclin-dependent kinase 5 regulates numerous neuronal functions with its activator, p35. Under neurotoxic conditions, p35 undergoes proteolytic cleavage to liberate p25, which has been implicated in various neurodegenerative diseases. Here, we show that p25 is generated following neuronal activity under physiological conditions in a GluN2B- and CaMKIIα-dependent manner. Moreover, we developed a knockin mouse model in which endogenous p35 is replaced with a calpain-resistant mutant p35 (Δp35KI) to prevent p25 generation. The Δp35KI mice exhibit impaired long-term depression and defective memory extinction, likely mediated through persistent GluA1 phosphorylation at Ser845. Finally, crossing the Δp35KI mice with the 5XFAD mouse model of Alzheimer's disease (AD) resulted in an amelioration of ß-amyloid (Aß)-induced synaptic depression and cognitive impairment. Together, these results reveal a physiological role of p25 production in synaptic plasticity and memory and provide new insights into the function of p25 in Aß-associated neurotoxicity and AD-like pathology.


Assuntos
Doença de Alzheimer/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Calpaína/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cognição , Quinase 5 Dependente de Ciclina/metabolismo , Modelos Animais de Doenças , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Endocitose , Técnicas de Introdução de Genes , Hipocampo/metabolismo , Humanos , Potenciação de Longa Duração , Depressão Sináptica de Longo Prazo , Camundongos , Proteínas do Tecido Nervoso/genética , Fosfotransferases , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses
9.
Science ; 342(6161): 983-7, 2013 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-24158904

RESUMO

The induced pluripotent stem (iPS) cell field holds promise for in vitro disease modeling. However, identifying innate cellular pathologies, particularly for age-related neurodegenerative diseases, has been challenging. Here, we exploited mutation correction of iPS cells and conserved proteotoxic mechanisms from yeast to humans to discover and reverse phenotypic responses to α-synuclein (αsyn), a key protein involved in Parkinson's disease (PD). We generated cortical neurons from iPS cells of patients harboring αsyn mutations, who are at high risk of developing PD dementia. Genetic modifiers from unbiased screens in a yeast model of αsyn toxicity led to identification of early pathogenic phenotypes in patient neurons. These included nitrosative stress, accumulation of endoplasmic reticulum (ER)-associated degradation substrates, and ER stress. A small molecule identified in a yeast screen (NAB2), and the ubiquitin ligase Nedd4 it affects, reversed pathologic phenotypes in these neurons.


Assuntos
Benzimidazóis/farmacologia , Neurônios/efeitos dos fármacos , Doença de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Animais , Benzimidazóis/química , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Mutação , Neurogênese , Neurônios/metabolismo , Neurônios/patologia , Doença de Parkinson/genética , Ratos , alfa-Sinucleína/genética
10.
Neurodegener Dis ; 10(1-4): 191-4, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22301371

RESUMO

Epigenetic mechanisms regulate the interaction between the genome and the environment and have been implicated in the etiology of various brain diseases. One type of epigenetic modification, histone acetylation, is dynamically altered during memory formation. Histone acetylation is regulated by the activities of histone deacetylase (HDAC) and histone acetyltransferase enzymes. The use of HDAC inhibitors has emerged as a promising new strategy for the therapeutic intervention of neurodegenerative disease. We used a combination of pharmacological and mouse genetic approaches that allowed us to identify HDAC2 as a specific negative regulator of synaptic plasticity and memory formation. Our results suggest that HDAC inhibitors enhance cognitive function by inhibiting HDAC2, which renders HDAC2 target genes more accessible to transcriptional activators and coactivators recruited by neuronal activity stimulation. The data presented at the 2011 Barcelona ADPD Conference delineate a novel and important role for HDAC2 activity in the cognitive impairments associated with neurodegenerative disease.


Assuntos
Transtornos Cognitivos/etiologia , Transtornos Cognitivos/genética , Epigênese Genética/genética , Doenças Neurodegenerativas/complicações , Doenças Neurodegenerativas/genética , Animais , Transtornos Cognitivos/tratamento farmacológico , Quinase 5 Dependente de Ciclina/genética , Modelos Animais de Doenças , Histona Desacetilase 2/genética , Inibidores de Histona Desacetilases/uso terapêutico , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Mutação/genética , Proteínas do Tecido Nervoso/genética , Fosfotransferases
11.
Neural Plast ; 2011: 537659, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21826278

RESUMO

Astrocytes have the ability to modulate neuronal excitability and synaptic transmission by the release of gliotransmitters. The importance of ATP released downstream of the activation of Gq-coupled receptors has been well established, but the mechanisms by which this release is regulated are unclear. The current work reveals that the elevation of diacylglycerol (DAG) in astrocytes induces vesicular ATP release. Unexpectedly, DAG-induced ATP release was found to be independent of PKC activation, but dependent upon activation of a C1 domain-containing protein. Astrocytes express the C1 domain-containing protein Munc13-1, which has been implicated in neuronal transmitter release, and RNAi-targeted downregulation of Munc13-1 inhibits astrocytic ATP release. These studies demonstrate that elevations of DAG induce the exocytotic release of ATP in astrocytes, likely via a Munc13-1-dependent mechanism.


Assuntos
Trifosfato de Adenosina/metabolismo , Astrócitos/metabolismo , Diglicerídeos/metabolismo , Transdução de Sinais , Animais , Animais Recém-Nascidos , Western Blotting , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Exocitose/fisiologia , Genes Reporter , Imuno-Histoquímica , Indicadores e Reagentes , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Proteínas do Tecido Nervoso/genética , Proteína Quinase C/metabolismo , RNA Interferente Pequeno/farmacologia , Receptores Purinérgicos P1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transmissão Sináptica/fisiologia
12.
Endocrinology ; 152(6): 2353-63, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21486931

RESUMO

We previously identified synaptic cell adhesion molecule 1 (SynCAM1) as a component of a genetic network involved in the hypothalamic control of female puberty. Although it is well established that SynCAM1 is a synaptic adhesion molecule, its contribution to hypothalamic function is unknown. Here we show that, in addition to the expected neuronal localization illustrated by its presence in GnRH neurons, SynCAM1 is expressed in hypothalamic astrocytes. Cell adhesion assays indicated that SynCAM is recognized by both GnRH neurons and astrocytes as an adhesive partner and promotes cell-cell adhesiveness via homophilic, extracellular domain-mediated interactions. Alternative splicing of the SynCAM1 primary mRNA transcript yields four mRNAs encoding membrane-spanning SynCAM1 isoforms. Variants 1 and 4 are predicted to be both N and O glycosylated. Hypothalamic astrocytes and GnRH-producing GT1-7 cells express mainly isoform 4 mRNA, and sequential N- and O-deglycosylation of proteins extracted from these cells yields progressively smaller SynCAM1 species, indicating that isoform 4 is the predominant SynCAM1 variant expressed in astrocytes and GT1-7 cells. Neither cell type expresses the products of two other SynCAM genes (SynCAM2 and SynCAM3), suggesting that SynCAM-mediated astrocyte-astrocyte and astrocyte-GnRH neuron adhesiveness is mostly mediated by SynCAM1 homophilic interactions. When erbB4 receptor function is disrupted in astrocytes, via transgenic expression of a dominant-negative erbB4 receptor form, SynCAM1-mediated adhesiveness is severely compromised. Conversely, SynCAM1 adhesive behavior is rapidly, but transiently, enhanced in astrocytes by ligand-dependent activation of erbB4 receptors, suggesting that erbB4-mediated events affecting SynCAM1 function contribute to regulate astrocyte adhesive communication.


Assuntos
Astrócitos/citologia , Moléculas de Adesão Celular/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/citologia , Hipotálamo/metabolismo , Imunoglobulinas/metabolismo , Neurônios/citologia , Sequência de Aminoácidos , Animais , Astrócitos/metabolismo , Adesão Celular , Molécula 1 de Adesão Celular , Moléculas de Adesão Celular/genética , Comunicação Celular , Linhagem Celular , Feminino , Imunoglobulinas/genética , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Neurônios/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transdução de Sinais
13.
Endocrinology ; 152(6): 2364-76, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21486934

RESUMO

Female sexual maturation requires erythroblastosis B (erbB)4 signaling in hypothalamic astrocytes; however, the mechanisms by which erbB4 contributes to this process are incompletely understood. Here we show that SynCAM1, a synaptic adhesion molecule with signaling capabilities, is not only expressed highly in neurons, but also in hypothalamic astrocytes and is functionally associated with erbB4 receptor activity. Whereas SynCAM1 expression is diminished in astrocytes with impaired erbB4 signaling, ligand-dependent activation of astroglial erbB4 receptors results in rapid association of erbB4 with SynCAM1 and activation of SynCAM1 gene transcription. To determine whether astrocytic SynCAM1-dependent intracellular signaling is required for normal female reproductive function, we generated transgenic mice that express in an astrocyte-specific manner a dominant-negative form of SynCAM1 lacking the intracellular domain. The mutant protein was correctly targeted to the cell membrane and was functionally viable as shown by its ability to block intracellular calcium/calmodulin-dependent serine protein kinase redistribution, a major SynCAM1-mediated event. Dominant-negative-SynCAM1 female mice had a delayed onset of puberty, disrupted estrous cyclicity, and reduced fecundity. These deficits were associated with a reduced capacity of neuregulin-dependent erbB4 receptor activation to elicit prostaglandin E2 release from astrocytes and GnRH release from the hypothalamus. We conclude that one of the mechanisms underlying erbB4 receptor-mediated facilitation of glial-neuronal interactions in the neuroendocrine brain involves SynCAM1-dependent signaling and that this interaction is required for normal female reproductive function.


Assuntos
Astrócitos/metabolismo , Receptores ErbB/metabolismo , Camundongos/metabolismo , Desenvolvimento Sexual , Animais , Astrócitos/citologia , Encéfalo/citologia , Encéfalo/metabolismo , Dinoprostona/metabolismo , Receptores ErbB/genética , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Camundongos/genética , Camundongos/crescimento & desenvolvimento , Camundongos Transgênicos , Ligação Proteica , Receptor ErbB-4 , Transdução de Sinais
14.
Endocrinology ; 148(11): 5147-61, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17615149

RESUMO

Much has been learned in recent years about the central mechanisms controlling the initiation of mammalian puberty. It is now clear that this process requires the interactive participation of several genes. Using a combination of high throughput, molecular, and bioinformatics strategies, in combination with a system biology approach, we singled out from the hypothalamus of nonhuman primates and rats a group of related genes whose expression increases at the time of female puberty. Although these genes [henceforth termed tumor-related genes (TRGs)] have diverse cellular functions, they share the common feature of having been earlier identified as involved in tumor suppression/tumor formation. A prominent member of this group is KiSS1, a gene recently shown to be essential for the occurrence of puberty. Cis-regulatory analysis revealed the presence of a hierarchically arranged gene set containing five major hubs (CDP/CUTL1, MAF, p53, YY1, and USF2) controlling the network at the transcriptional level. In turn, these hubs are heavily connected to non-TRGs involved in the transcriptional regulation of the pubertal process. TRGs may be expressed in the mammalian hypothalamus as components of a regulatory gene network that facilitates and integrates cellular and cell-cell communication programs required for the acquisition of female reproductive competence.


Assuntos
Redes Reguladoras de Genes , Genes Neoplásicos , Hipotálamo/metabolismo , Maturidade Sexual/genética , Regulação para Cima , Região 5'-Flanqueadora , Animais , Sítios de Ligação , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Macaca mulatta , Modelos Biológicos , Sistemas Neurossecretores/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição/metabolismo
15.
J Neurosci ; 26(51): 13167-79, 2006 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-17182767

RESUMO

Thyroid transcription factor 1 (TTF1) [also known as Nkx2.1 (related to the NK-2 class of homeobox genes) and T/ebp (thyroid-specific enhancer-binding protein)], a homeodomain gene required for basal forebrain morphogenesis, remains expressed in the hypothalamus after birth, suggesting a role in neuroendocrine function. Here, we show an involvement of TTF1 in the control of mammalian puberty and adult reproductive function. Gene expression profiling of the nonhuman primate hypothalamus revealed that TTF1 expression increases at puberty. Mice in which the Ttf1 gene was ablated from differentiated neurons grew normally and had normal basal ganglia/hypothalamic morphology but exhibited delayed puberty, reduced reproductive capacity, and a short reproductive span. These defects were associated with reduced hypothalamic expression of genes required for sexual development and deregulation of a gene involved in restraining puberty. No extrapyramidal impairments associated with basal ganglia dysfunction were apparent. Thus, although TTF1 appears to fulfill only a morphogenic function in the ventral telencephalon, once this function is satisfied in the hypothalamus, TTF1 remains active as part of the transcriptional machinery controlling female sexual development.


Assuntos
Gânglios da Base/fisiologia , Diferenciação Celular/genética , Deleção de Genes , Neurônios/citologia , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Reprodução/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Animais , Gânglios da Base/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Hipotálamo/citologia , Hipotálamo/fisiologia , Macaca mulatta , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neurônios/fisiologia , Proteínas Nucleares/deficiência , Comportamento Sexual Animal/fisiologia , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/deficiência
16.
Mol Cell Endocrinol ; 254-255: 51-9, 2006 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-16753258

RESUMO

Mammalian puberty requires activation of luteinizing hormone-releasing hormone (LHRH) neurons. In turn, these neurons are controlled by transsynaptic and glia-to-neuron communication pathways, which employ diverse cellular proteins for proper function. We have now used a high throughput relative quantitative proteomics technique to identify such proteins. We selected the method of two-dimensional liquid chromatography tandem mass spectrometry (2DLC-MS/MS) and cleavable isotope-coded affinity tags (cICAT), to both identify and quantify individual proteins within a complex protein mixture. The proteins used derived from the hypothalamus of juvenile (25-day-old) and peripubertal (first proestrus, LP) female rats, and their identity was established by analyzing their mass spectra via database searching. Five proteins involved in glutamate metabolism were detected and two of them appeared to be differentially expressed. They were selected for further analysis, because of their importance in controlling glutamate synthesis and degradation, and their preferential expression in astroglial cells. One, glutamate dehydrogenase (GDH) catalyzes glutamate synthesis; its hypothalamic content detected by 2DLC-MS/MS increases at first proestrus. The other, glutamine synthetase (GS), catalyzes the metabolism of glutamate to glutamine; its content decreases in proestrus. Western blot analysis verified these results. Because these changes suggested an increased glutamate production at puberty, we measured glutamate release from hypothalamic fragments from juvenile 29-day old rats, and from rats treated with PMSG to induce a premature proestrus surge of luteinizing hormone (LH). To determine the net output of glutamate in the absence of re-uptake we used the excitatory amino acid transporter (EAAT) inhibitor l-trans-pyrrolidine-2,4-dicarboxylic acid (PDC). PDC elicited significantly more glutamate- and LHRH-release from the proestrus hypothalamus. Thus, an increase excitatory drive to the LHRH neuronal network provided by glutamatergic inputs of glial origin, is an event contributing to the pubertal activation of LHRH secretion.


Assuntos
Fatores Etários , Ácido Glutâmico/metabolismo , Neuroglia/metabolismo , Proteômica/métodos , Maturidade Sexual , Animais , Feminino , Perfilação da Expressão Gênica , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas Equinas/farmacologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Modelos Biológicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley
17.
Endocrinology ; 147(3): 1166-74, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16373420

RESUMO

The initiation of mammalian puberty requires an increase in pulsatile release of GnRH from the hypothalamus. This increase is brought about by coordinated changes in transsynaptic and glial-neuronal communication. As the neuronal and glial excitatory inputs to the GnRH neuronal network increase, the transsynaptic inhibitory tone decreases, leading to the pubertal activation of GnRH secretion. The excitatory neuronal systems most prevalently involved in this process use glutamate and the peptide kisspeptin for neurotransmission/neuromodulation, whereas the most important inhibitory inputs are provided by gamma-aminobutyric acid (GABA)ergic and opiatergic neurons. Glial cells, on the other hand, facilitate GnRH secretion via growth factor-dependent cell-cell signaling. Coordination of this regulatory neuronal-glial network may require a hierarchical arrangement. One level of coordination appears to be provided by a host of unrelated genes encoding proteins required for cell-cell communication. A second, but overlapping, level might be provided by a second tier of genes engaged in specific cell functions required for productive cell-cell interaction. A third and higher level of control involves the transcriptional regulation of these subordinate genes by a handful of upper echelon genes that, operating within the different neuronal and glial subsets required for the initiation of the pubertal process, sustain the functional integration of the network. The existence of functionally connected genes controlling the pubertal process is consistent with the concept that puberty is under genetic control and that the genetic underpinnings of both normal and deranged puberty are polygenic rather than specified by a single gene. The availability of improved high-throughput techniques and computational methods for global analysis of mRNAs and proteins will allow us to not only initiate the systematic identification of the different components of this neuroendocrine network but also to define their functional interactions.


Assuntos
Sistema Endócrino/fisiologia , Neurônios/metabolismo , Sistemas Neurossecretores , Puberdade , Biologia de Sistemas/métodos , Animais , Comunicação Celular , DNA/metabolismo , Genes Supressores de Tumor , Hormônio Liberador de Gonadotropina/metabolismo , Substâncias de Crescimento/metabolismo , Humanos , Hipotálamo/metabolismo , Kisspeptinas , Modelos Biológicos , Neuroglia/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas , RNA Mensageiro/metabolismo , Transdução de Sinais , Software , Sinapses , Transcrição Gênica , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...