Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Front Mol Neurosci ; 15: 932497, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35909451

RESUMO

This article reviews the current progress in our understanding of the mechanisms by which growth factors, including brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF), and select neurotrophin-regulated gene products, such as VGF (non-acronymic) and VGF-derived neuropeptides, function in the central nervous system (CNS) to modulate neuropsychiatric and neurodegenerative disorders, with a discussion of the possible therapeutic applications of these growth factors to major depressive disorder (MDD) and Alzheimer's disease (AD). BDNF and VEGF levels are generally decreased regionally in the brains of MDD subjects and in preclinical animal models of depression, changes that are associated with neuronal atrophy and reduced neurogenesis, and are reversed by conventional monoaminergic and novel ketamine-like antidepressants. Downstream of neurotrophins and their receptors, VGF was identified as a nerve growth factor (NGF)- and BDNF-inducible secreted protein and neuropeptide precursor that is produced and trafficked throughout the CNS, where its expression is greatly influenced by neuronal activity and exercise, and where several VGF-derived peptides modulate neuronal activity, function, proliferation, differentiation, and survival. Moreover, levels of VGF are reduced in the CSF of AD subjects, where it has been repetitively identified as a disease biomarker, and in the hippocampi of subjects with MDD, suggesting possible shared mechanisms by which reduced levels of VGF and other proteins that are similarly regulated by neurotrophin signaling pathways contribute to and potentially drive the pathogenesis and progression of co-morbid neuropsychiatric and neurodegenerative disorders, particularly MDD and AD, opening possible therapeutic windows.

2.
J Biol Chem ; 291(40): 21322-21334, 2016 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-27466366

RESUMO

Reproductive function is controlled by the pulsatile release of hypothalamic gonadotropin-releasing hormone (GnRH), which regulates the expression of the gonadotropins luteinizing hormone and FSH in pituitary gonadotropes. Paradoxically, Fshb gene expression is maximally induced at lower frequency GnRH pulses, which provide a very low average concentration of GnRH stimulation. We studied the role of secreted factors in modulating gonadotropin gene expression. Inhibition of secretion specifically disrupted gonadotropin subunit gene regulation but left early gene induction intact. We characterized the gonadotrope secretoproteome and global mRNA expression at baseline and after Gαs knockdown, which has been found to increase Fshb gene expression (1). We identified 1077 secreted proteins or peptides, 19 of which showed mRNA regulation by GnRH or/and Gαs knockdown. Among several novel secreted factors implicated in Fshb gene regulation, we focused on the neurosecretory protein VGF. Vgf mRNA, whose gene has been implicated in fertility (2), exhibited high induction by GnRH and depended on Gαs In contrast with Fshb induction, Vgf induction occurred preferentially at high GnRH pulse frequency. We hypothesized that a VGF-derived peptide might regulate Fshb gene induction. siRNA knockdown or extracellular immunoneutralization of VGF augmented Fshb mRNA induction by GnRH. GnRH stimulated the secretion of the VGF-derived peptide NERP1. NERP1 caused a concentration-dependent decrease in Fshb gene induction. These findings implicate a VGF-derived peptide in selective regulation of the Fshb gene. Our results support the concept that signaling specificity from the cell membrane GnRH receptor to the nuclear Fshb gene involves integration of intracellular signaling and exosignaling regulatory motifs.


Assuntos
Hormônio Foliculoestimulante/biossíntese , Regulação da Expressão Gênica/fisiologia , Gonadotrofos/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neuropeptídeos/metabolismo , Peptídeos/metabolismo , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Gonadotrofos/citologia , Camundongos , Fatores de Crescimento Neural , RNA Mensageiro/biossíntese
3.
Endocrinology ; 157(3): 983-96, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26730934

RESUMO

Environmental enrichment (EE), a housing condition providing complex physical, social, and cognitive stimulation, leads to improved metabolic health and resistance to diet-induced obesity and cancer. One underlying mechanism is the activation of the hypothalamic-sympathoneural-adipocyte axis with hypothalamic brain-derived neurotrophic factor (BDNF) as the key mediator. VGF, a peptide precursor particularly abundant in the hypothalamus, was up-regulated by EE. Overexpressing BDNF or acute injection of BDNF protein to the hypothalamus up-regulated VGF, whereas suppressing BDNF signaling down-regulated VGF expression. Moreover, hypothalamic VGF expression was regulated by leptin, melanocortin receptor agonist, and food deprivation mostly paralleled to BDNF expression. Recombinant adeno-associated virus-mediated gene transfer of Cre recombinase to floxed VGF mice specifically decreased VGF expression in the hypothalamus. In contrast to the lean and hypermetabolic phenotype of homozygous germline VGF knockout mice, specific knockdown of hypothalamic VGF in male adult mice led to increased adiposity, decreased core body temperature, reduced energy expenditure, and impaired glucose tolerance, as well as disturbance of molecular features of brown and white adipose tissues without effects on food intake. However, VGF knockdown failed to block the EE-induced BDNF up-regulation or decrease of adiposity indicating a minor role of VGF in the hypothalamic-sympathoneural-adipocyte axis. Taken together, our results suggest hypothalamic VGF responds to environmental demands and plays an important role in energy balance and glycemic control likely acting in the melanocortin pathway downstream of BDNF.


Assuntos
Adaptação Fisiológica/genética , Adipócitos/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Metabolismo Energético/genética , Meio Ambiente , Hipotálamo/metabolismo , Neuropeptídeos/genética , Obesidade/genética , Sistema Nervoso Simpático/metabolismo , Adaptação Fisiológica/efeitos dos fármacos , Adipócitos/efeitos dos fármacos , Adiposidade , Animais , Glicemia/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Metabolismo Energético/efeitos dos fármacos , Privação de Alimentos , Hipotálamo/efeitos dos fármacos , Leptina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Crescimento Neural , Neuropeptídeos/efeitos dos fármacos , Receptores de Melanocortina/agonistas , Meio Social , Sistema Nervoso Simpático/efeitos dos fármacos , Regulação para Cima
4.
Endocr Rev ; 32(6): 755-97, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21862681

RESUMO

The chromogranins (chromogranin A and chromogranin B), secretogranins (secretogranin II and secretogranin III), and additional related proteins (7B2, NESP55, proSAAS, and VGF) that together comprise the granin family subserve essential roles in the regulated secretory pathway that is responsible for controlled delivery of peptides, hormones, neurotransmitters, and growth factors. Here we review the structure and function of granins and granin-derived peptides and expansive new genetic evidence, including recent single-nucleotide polymorphism mapping, genomic sequence comparisons, and analysis of transgenic and knockout mice, which together support an important and evolutionarily conserved role for these proteins in large dense-core vesicle biogenesis and regulated secretion. Recent data further indicate that their processed peptides function prominently in metabolic and glucose homeostasis, emotional behavior, pain pathways, and blood pressure modulation, suggesting future utility of granins and granin-derived peptides as novel disease biomarkers.


Assuntos
Cromograninas/química , Cromograninas/fisiologia , Animais , Biomarcadores/química , Biomarcadores/metabolismo , Cromograninas/uso terapêutico , Células Endócrinas/efeitos dos fármacos , Células Endócrinas/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/química , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/uso terapêutico , Humanos , Fatores de Crescimento Neural/química , Fatores de Crescimento Neural/fisiologia , Fatores de Crescimento Neural/uso terapêutico , Células Neuroendócrinas/efeitos dos fármacos , Células Neuroendócrinas/metabolismo , Proteína Secretora Neuroendócrina 7B2/química , Proteína Secretora Neuroendócrina 7B2/fisiologia , Proteína Secretora Neuroendócrina 7B2/uso terapêutico , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeos/química , Neuropeptídeos/fisiologia , Neuropeptídeos/uso terapêutico , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/fisiologia , Fragmentos de Peptídeos/uso terapêutico , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Homologia de Sequência de Aminoácidos
5.
J Neurosci ; 28(39): 9857-69, 2008 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-18815270

RESUMO

VGF is a neurotrophin-inducible, activity-regulated gene product that is expressed in CNS and PNS neurons, in which it is processed into peptides and secreted. VGF synthesis is stimulated by BDNF, a critical regulator of hippocampal development and function, and two VGF C-terminal peptides increase synaptic activity in cultured hippocampal neurons. To assess VGF function in the hippocampus, we tested heterozygous and homozygous VGF knock-out mice in two different learning tasks, assessed long-term potentiation (LTP) and depression (LTD) in hippocampal slices from VGF mutant mice, and investigated how VGF C-terminal peptides modulate synaptic plasticity. Treatment of rat hippocampal slices with the VGF-derived peptide TLQP62 resulted in transient potentiation through a mechanism that was selectively blocked by the BDNF scavenger TrkB-Fc, the Trk tyrosine kinase inhibitor K252a (100 nm), and tPA STOP, an inhibitor of tissue plasminogen activator (tPA), an enzyme involved in pro-BDNF cleavage to BDNF, but was not blocked by the NMDA receptor antagonist APV, anti-p75(NTR) function-blocking antiserum, or previous tetanic stimulation. Although LTP was normal in slices from VGF knock-out mice, LTD could not be induced, and VGF mutant mice were impaired in hippocampal-dependent spatial learning and contextual fear conditioning tasks. Our studies indicate that the VGF C-terminal peptide TLQP62 modulates hippocampal synaptic transmission through a BDNF-dependent mechanism and that VGF deficiency in mice impacts synaptic plasticity and memory in addition to depressive behavior.


Assuntos
Comportamento Animal/fisiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hipocampo/citologia , Potenciação de Longa Duração/fisiologia , Neuropeptídeos/fisiologia , Análise de Variância , Animais , Condicionamento Clássico/fisiologia , Relação Dose-Resposta à Radiação , Estimulação Elétrica/métodos , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Medo , Técnicas In Vitro , Deficiências da Aprendizagem/genética , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/efeitos da radiação , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/fisiologia , Fatores de Crescimento Neural , Neuropeptídeos/deficiência , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Valina/análogos & derivados , Valina/farmacologia
6.
J Comp Neurol ; 510(4): 351-66, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18651636

RESUMO

Axonal growth cones initiate and sustain directed growth in response to cues in their environment. A variety of events such as receptor internalization, kinase activation, and actin rearrangement can be stimulated by guidance cues and are essential for mediating targeted growth cone behavior. Surprisingly little is known about how such disparate actions are coordinated. Our data suggest that ezrin, radixin, and moesin (ERMs), a family of highly homologous, multifunctional proteins may be able to coordinate growth cone responses to the guidance cue Semaphorin 3A (Sema3A). We show that active ERMs concentrate asymmetrically in neocortical growth cones, are rapidly and transiently inactivated by Sema3A, and are required for Sema3A-mediated growth cone collapse and guidance. The FERM domain of active ERMs regulates internalization of the Sema3A receptor, Npn1, and its coreceptor, L1CAM, while the ERM C-terminal domain binds and caps F-actin. Our data support a model in which ERMs can coordinate membrane and actin dynamics in response to Sema3A.


Assuntos
Axônios/fisiologia , Proteínas de Ligação a DNA/metabolismo , Cones de Crescimento/fisiologia , Semaforina-3A/metabolismo , Fatores de Transcrição/metabolismo , Actinas/fisiologia , Animais , Galinhas , Proteínas do Citoesqueleto/genética , Proteínas de Ligação a DNA/genética , Camundongos , Mutação , Plasmídeos , Coelhos , Receptores de Superfície Celular/fisiologia , Semaforina-3A/genética , Fatores de Transcrição/genética
7.
Brain Res ; 1217: 86-95, 2008 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-18502406

RESUMO

Circulating gonadal steroid levels affect metabolic homeostasis by regulating appetite and food intake. The actions of estrogen are mediated through its two receptors ERalpha and ERbeta. ERalpha expression is necessary to maintain normal food intake, body weight and adiposity. Leptin plays a central role in regulating feeding behavior, homeostasis and reproduction. It is known that there is an effect of estrogen and leptin on feeding behavior. The present study was undertaken 1) to assess the changes in the reproductive cycle in obese, infertile ob/ob mice with no circulating leptin and infertile, obese, agouti (Ay/a) mice with high circulating leptin levels, 2) to evaluate the hypothalamic distribution of ERalpha and ERbeta, and 3) to analyze the differences in expression of ERs related to leptin and beta-estradiol levels in these mouse lines. The results show that the ob/ob and Ay/a mice were acyclic and were at a persistent estrous phase. The beta-estradiol levels were similar between WT, ob/ob and Ay/a mice. Stereologic analysis showed that there were significantly higher numbers of ERalpha-immunoreactive cells in ob/ob mice irrespective of sex when compared to wild-type (WT) in arcuate nucleus (ARH) and no significant change in ERbeta immunoreactive cell numbers in ARH or paraventricular nucleus (PVN). Ovariectomy in female wild-type mice caused a 50% increase of ERalpha-immunoreactive cells. Results suggest that leptin and estrogen act via the same neuronal circuits to affect reproduction, neuroendocrine and behavioral processes. However, estrogen levels and acyclicity have more profound effect on the regulation of ERalpha cell numbers in the ARH than circulating leptin levels.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Ciclo Estral/fisiologia , Obesidade/metabolismo , Receptores de Estrogênio/biossíntese , Animais , Estradiol/metabolismo , Feminino , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Leptina/metabolismo , Masculino , Camundongos , Camundongos Obesos , Ovariectomia
8.
Brain Res ; 1089(1): 21-32, 2006 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-16631141

RESUMO

Two novel granin-like polypeptides, VGF and pro-SAAS, which are stored in and released from secretory vesicles and are expressed widely in nervous, endocrine, and neuroendocrine tissues, play roles in the regulation of body weight, feeding, and energy expenditure. Both VGF and pro-SAAS are cleaved into peptide fragments, several of which are biologically active. We utilized a highly sensitive and specific radioimmunoassay (RIA) to immunoreactive, pro-SAAS-derived PEN peptides, developed another against immunoreactive, VGF-derived AQEE30 peptides, and quantified these peptides in various mouse tissues and brain regions. Immunoreactive AQEE30 was most abundant in the pituitary, while brain levels were highest in hypothalamus, striatum, and frontal cortex. Immunoreactive PEN levels were highest in the pancreas and spinal cord, and in brain, PEN was most abundant in striatum, hippocampus, pons and medulla, and cortex. Since both peptides were expressed in hypothalamus, a region of the brain that controls feeding and energy expenditure, double label immunofluorescence studies were employed. These demonstrated that 42% of hypothalamic arcuate neurons coexpress VGF and SAAS peptides, and that the intracellular distributions of these peptides in arcuate neurons differed. By RIA, cold stress increased immunoreactive AQEE30 and PEN peptide levels in female but not male hypothalamus, while a high fat diet increased AQEE30 and PEN peptide levels in female but not male hippocampus. VGF and SAAS-derived peptides are therefore widely expressed in endocrine, neuroendocrine, and neural tissues, can be accurately quantified by RIA, and are differentially regulated in the brain by diet and cold stress.


Assuntos
Encéfalo/metabolismo , Ingestão de Energia/fisiologia , Comportamento Alimentar/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/metabolismo , Estresse Fisiológico/metabolismo , Animais , Encéfalo/anatomia & histologia , Encéfalo/fisiopatologia , Temperatura Baixa/efeitos adversos , Gorduras na Dieta/efeitos adversos , Modelos Animais de Doenças , Feminino , Alimentos Formulados/efeitos adversos , Hipocampo/anatomia & histologia , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Hipotálamo/anatomia & histologia , Hipotálamo/metabolismo , Hipotálamo/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural , Proteínas do Tecido Nervoso/química , Neurônios/metabolismo , Neuropeptídeos/química , Especificidade de Órgãos , Pâncreas/metabolismo , Pâncreas/fisiopatologia , Peptídeos/análise , Peptídeos/metabolismo , Radioimunoensaio/efeitos adversos , Caracteres Sexuais , Estresse Fisiológico/etiologia , Estresse Fisiológico/fisiopatologia , Regulação para Cima/fisiologia
9.
J Biol Chem ; 280(50): 41595-608, 2005 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-16221685

RESUMO

Distinct intracellular pathways are involved in regulated and constitutive protein secretion from neuronal and endocrine cells, yet the peptide signals and molecular mechanisms responsible for targeting and retention of soluble proteins in secretory granules are incompletely understood. By using confocal microscopy and subcellular fractionation, we examined trafficking of the neuronal and endocrine peptide precursor VGF that is stored in large dense core vesicles and undergoes regulated secretion. VGF cofractionated with secretory vesicle membranes but was not detected in detergent-resistant lipid rafts. Deletional analysis using epitope-tagged VGF suggested that the C-terminal 73-amino acid fragment of VGF, containing two predicted alpha-helical loops and four potential prohormone convertase (PC) cleavage sites, was necessary and sufficient with an N-terminal signal peptide-containing domain, for large dense core vesicle sorting and regulated secretion from PC12 and INS-1 cells. Further transfection analysis identified the sorting sequence as a compact C-terminal alpha-helix and embedded 564RRR566 PC cleavage site; mutation of the 564RRR566 PC site in VGF-(1-65): GFP:VGF-(545-617) blocked regulated secretion, whereas disruption of the alpha-helix had no effect. Mutation of the adjacent 567HFHH570 motif, a charged region that might enhance PC cleavage in acidic environments, also blocked regulated release. Finally, inhibition of PC cleavage in PC12 cells using the membrane-permeable synthetic peptide chloromethyl ketone (decanoyl-RVKR-CMK) blocked regulated secretion of VGF. Our studies define a critical RRR-containing C-terminal domain that targets VGF into the regulated pathway in neuronal PC12 and endocrine INS-1 cells, providing additional support for the proposed role that PCs and their cleavage sites play in regulated peptide secretion.


Assuntos
Regulação da Expressão Gênica , Neurônios/metabolismo , Peptídeos/química , Pró-Proteína Convertases/química , Proteínas/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Western Blotting , Linhagem Celular , Membrana Celular/metabolismo , Centrifugação com Gradiente de Concentração , Cromograninas/química , Detergentes/farmacologia , Epitopos/química , Deleção de Genes , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/metabolismo , Lipídeos/química , Microdomínios da Membrana/química , Microscopia Confocal , Modelos Genéticos , Dados de Sequência Molecular , Mutação , Neuropeptídeos , Células PC12 , Plasmídeos/metabolismo , Sinais Direcionadores de Proteínas , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes de Fusão/química , Transdução de Sinais , Frações Subcelulares/metabolismo , Fatores de Tempo , Transfecção
10.
Endocrinology ; 146(12): 5151-63, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16141392

RESUMO

Targeted deletion of the gene encoding the neuronal and endocrine secreted peptide precursor called VGF (nonacronymic) produces a lean, hypermetabolic, hyperactive mouse. Because VGF mutant mice are resistant to specific forms of diet-, lesion-, and genetically induced obesity, we investigated the role that this polypeptide plays in glucose homeostasis. We report that VGF mutant mice have increased insulin sensitivity by hyperinsulinemic euglycemic clamp analysis, and by insulin and glucose tolerance testing. Blunted counterregulatory responses in VGF-deficient mice were likely influenced by their significantly lower liver glycogen levels. VGF deficiency lowered circulating glucose and insulin levels in several murine models of obesity that are also susceptible to adult onset diabetes mellitus, including A(y)/a agouti, ob/ob, and MC4R(-)/MC4R(-) mice. Interestingly, ablation of Vgf in ob/ob mice decreased circulating glucose and insulin levels but did not affect adiposity, whereas MC4R(-)/MC4R(-) mice that are additionally deficient in VGF have improved insulin responsiveness at 7-8 wk of age, when lean MC4R(-)/MC4R(-) mice already have impaired insulin tolerance but are not yet obese. VGF mutant mice also resisted developing obesity and hyperglycemia in response to a high-fat/high-carbohydrate diet, and after gold thioglucose treatment, which is toxic to hypothalamic glucose-sensitive neurons. Lastly, circulating adiponectin, an adipose-synthesized protein the levels of which are correlated with improved insulin sensitivity, increased in VGF mutant compared with wild-type mice. Modulation of VGF levels and/or VGF signaling may consequently represent an alternative means to regulate circulating glucose levels and insulin sensitivity.


Assuntos
Hiperglicemia/etiologia , Hiperinsulinismo/etiologia , Obesidade/complicações , Obesidade/metabolismo , Proteínas/metabolismo , Tecido Adiposo/metabolismo , Animais , Dieta/efeitos adversos , Modelos Animais de Doenças , Suscetibilidade a Doenças , Ingestão de Energia , Glucagon/metabolismo , Glucose/administração & dosagem , Glucose/metabolismo , Glucose/farmacocinética , Glucose/farmacologia , Homeostase , Hipoglicemia/induzido quimicamente , Hipoglicemia/fisiopatologia , Injeções Intraperitoneais , Insulina/metabolismo , Resistência à Insulina , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Crescimento Neural , Neuropeptídeos , Obesidade/genética , Proteínas/genética , Receptor Tipo 4 de Melanocortina/deficiência
11.
Cell Mol Neurobiol ; 24(4): 517-33, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15233376

RESUMO

1. The vgf gene encodes a neuropeptide precursor with a restricted pattern of expression that is limited to a subset of neurons in the central and peripheral nervous systems and to specific populations of endocrine cells in the adenohypophysis, adrenal medulla, gastrointestinal tract, and pancreas. In responsive neurons, vgf transcription is upregulated by neurotrophins. the basis for the original identification of VGF as nerve growth factor- (NGF) inducible in PC12 cells (A. Levi, J. D. Eldridge, and B. M. Paterson, Science 229:393-395, 1985). 2. In this review, we shall summarize data concerning the transcriptional regulation of vgf in vitro, the structural organization of the vgf promoter as well as the transcription factors which regulate its activity. 3. On the basis of in situ hybridization and immunohistochemical studies, the in vivo tissue-specific expression of VGF during differentiation and in the adult will be summarized. 4. Parallel biochemical data will be reviewed, addressing the proteolytical processing of the pro-VGF precursor within the secretory compartment of neuroendocrine cells. 5. Finally, analysis of the phenotype of VGF knockout mice will be discussed, implying a nonredundant role of VGF products in the regulation of energy storage and expenditure.


Assuntos
Células Cromafins/metabolismo , Neurônios/metabolismo , Neuropeptídeos/biossíntese , Sistemas Neurossecretores/metabolismo , Biossíntese de Proteínas , Animais , Metabolismo Energético/genética , Genes Reguladores/genética , Humanos , Especificidade de Órgãos , Regiões Promotoras Genéticas/genética , Proteínas/genética , Fatores de Transcrição/genética
12.
Neurochem Res ; 29(6): 1093-103, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15176466

RESUMO

DNA microarray analysis has been used to investigate relative changes in the level of gene expression in the CNS, including changes that are associated with disease, injury, psychiatric disorders, drug exposure or withdrawal, and memory formation. We have used oligonucleotide microarrays to identify hypothalamic genes that respond to nutritional manipulation. In addition to commonly used microarray analysis based on criteria such as fold-regulation, we have also found that simply carrying out multiple t tests then sorting by P value constitutes a highly reliable method to detect true regulation, as assessed by real-time polymerase chain reaction (PCR), even for relatively low abundance genes or relatively low magnitude of regulation. Such analyses directly suggested novel mechanisms that mediate effects of nutritional state on neuroendocrine function and are being used to identify regulated gene products that may elucidate the metabolic pathology of obese ob/ob, lean Vgf-/Vgf-, and other models with profound metabolic impairments.


Assuntos
Regulação da Expressão Gênica/genética , Hipotálamo/fisiologia , Metabolismo/genética , Fenômenos Fisiológicos da Nutrição , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Fenômenos Fisiológicos da Nutrição Animal , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL
13.
J Comp Neurol ; 464(4): 438-48, 2003 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-12900915

RESUMO

L1 is a member of the Ig superfamily of cell adhesion molecules (CAMs) that functions in many aspects of neuronal development including axonal outgrowth and neuronal migration. These functions require coordination between L1 and the actin cytoskeleton. Because CAMs and the cytoskeleton do not bind directly, membrane-cytoskeletal linkers (MCLs) such as ankyrin are thought to be crucial to their interactions, but data from a knockout mouse suggest that ankyrin is not necessary for the earliest events attributed to L1 function. Recent findings in hippocampal cell culture show that members of the ERM family of proteins (ezrin, radixin, and moesin) can also serve as MCLs between L1 and actin in neurons. Here, we demonstrate that ERM proteins are expressed in extending neuronal processes in the intermediate zone of the developing cortex, a region that is densely packed with migrating neurons and growing axons. ERMs and L1 are codistributed extensively over a transient time course that coincides with rapid axon growth and cortical expansion. This codistribution is strong at embryonic day 17 and 19 but diminishes by postnatal day 0, at which time ankyrin-L1 codistribution increases dramatically. These findings suggest that in the developing neocortex, ERMs are the predominant MCL for L1 during migration and axon extension, neither of which requires ankyrin function. Furthermore, these data suggest that there is a developmentally regulated switch in MCL function in the developing brain.


Assuntos
Proteínas Sanguíneas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Neocórtex/crescimento & desenvolvimento , Neocórtex/metabolismo , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Fosfoproteínas/metabolismo , Animais , Anquirinas/metabolismo , Axônios/fisiologia , Western Blotting , Feminino , Imuno-Histoquímica , Masculino , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
14.
Mt Sinai J Med ; 70(2): 93-100, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12634901

RESUMO

Neurotrophic growth factors are proteins that control neuronal differentiation and survival, and consequently play important roles in the developing and adult stages of the nervous system. Study of the genes that are regulated by these growth factors has provided insight into the proteins that are critical to the maturation of the nervous system, suggesting that select neurotrophins may play a role in the control of body homeostasis by the brain and peripheral nervous system. Our understanding of the mechanisms of action of neurotrophic growth factors has increased through experimental manipulation of cultured neurons and neuronal cell lines. In particular, the PC12 pheochromocytoma cell line, which displays many properties of adrenal chromaffin cells and undergoes differentiation into sympathetic neuron-like cells when treated with nerve growth factor, has been extensively investigated to identify components of neurotrophin signaling pathways as well as the genes that they regulate. VGF was one of the first neurotrophin-regulated clones identified in NGF-treated PC12 cells. Subsequent studies indicate that the vgf gene is regulated in vivo in the nervous system by neurotrophins, by electrical activity, in response to injury or seizure, and by feeding and the circadian clock. The vgf gene encodes a polypeptide rich in paired basic amino acids; this polypeptide is differentially processed in neuronal and neuroendocrine cells and is released via the regulated secretory pathway. Generation and analysis of knockout mice that fail to synthesize VGF indicate that this protein plays a critical, non-redundant role in the regulation of energy homeostasis, providing a possible link between neurotrophin function in the nervous system and the peripheral control of feeding and metabolic activity. Future experiments should clarify the sites and mechanisms of action of this neurotrophin-regulated neuronal and neuroendocrine protein.


Assuntos
Metabolismo Energético/fisiologia , Regulação da Expressão Gênica , Fatores de Crescimento Neural/fisiologia , Proteínas/genética , Animais , Camundongos , Camundongos Mutantes , Neuropeptídeos , Transdução de Sinais
15.
J Neurosci ; 22(16): 6929-38, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12177191

RESUMO

Targeted deletion of the gene encoding the neuronal and neuroendocrine secreted polypeptide VGF (nonacronymic) produces a lean, hypermetabolic mouse. Consistent with this phenotype, VGF mRNA levels are regulated in the hypothalamic arcuate nucleus in response to fasting. To gain insight into the site(s) and mechanism(s) of action of VGF, we further characterized VGF expression in the hypothalamus. Double-label studies indicated that VGF and pro-opiomelanocortin were coexpressed in lateral arcuate neurons in the fed state, and that VGF expression was induced after fasting in medial arcuate neurons that synthesize neuropeptide Y (NPY). Like NPY, VGF mRNA induction in this region of the hypothalamus in fasted mice was inhibited by exogenous leptin. In leptin-deficient ob/ob and receptor-mutant db/db mice, VGF mRNA levels in the medial arcuate were elevated. To identify neural pathways that are functionally compromised by Vgf ablation, VGF mutant mice were crossed with obese A(y)/a (agouti) and ob/ob mice. VGF deficiency completely blocked the development of obesity in A(y)/a mice, whereas deletion of Vgf in ob/ob mice attenuated weight gain but had no impact on adiposity. Hypothalamic levels of NPY and agouti-related polypeptide mRNAs in both double-mutant lines were dramatically elevated 10- to 15-fold above those of wild-type mice. VGF-deficient mice were also found to resist diet- and gold thioglucose-induced obesity. These data and the susceptibility of VGF mutant mice to monosodium glutamate-induced obesity are consistent with a role for VGF in outflow pathways, downstream of hypothalamic and/or brainstem melanocortin 4 receptors, that project via the autonomic nervous system to peripheral metabolic tissues and regulate energy homeostasis.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Dieta , Peptídeos e Proteínas de Sinalização Intercelular , Neurônios/metabolismo , Obesidade/metabolismo , Proteínas/metabolismo , Proteína Agouti Sinalizadora , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/patologia , Aurotioglucose , Modelos Animais de Doenças , Metabolismo Energético/genética , Jejum/metabolismo , Marcação de Genes , Imuno-Histoquímica , Hibridização In Situ , Leptina/farmacologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes , Fatores de Crescimento Neural , Vias Neurais/fisiopatologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neuropeptídeo Y/biossíntese , Neuropeptídeos , Obesidade/induzido quimicamente , Obesidade/patologia , Pró-Opiomelanocortina/biossíntese , Proteínas/genética , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Resposta de Saciedade , Glutamato de Sódio
16.
J Cell Biol ; 157(7): 1105-12, 2002 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-12070130

RESUMO

A yeast two-hybrid library was screened using the cytoplasmic domain of the axonal cell adhesion molecule L1 to identify binding partners that may be involved in the regulation of L1 function. The intracellular domain of L1 bound to ezrin, a member of the ezrin, radixin, and moesin (ERM) family of membrane-cytoskeleton linking proteins, at a site overlapping that for AP2, a clathrin adaptor. Binding of bacterial fusion proteins confirmed this interaction. To determine whether ERM proteins interact with L1 in vivo, extracellular antibodies to L1 were used to force cluster the protein on cultured hippocampal neurons and PC12 cells, which were then immunolabeled for ERM proteins. Confocal analysis revealed a precise pattern of codistribution between ERMs and L1 clusters in axons and PC12 neurites, whereas ERMs in dendrites and spectrin labeling remained evenly distributed. Transfection of hippocampal neurons grown on an L1 substrate with a dominant negative ERM construct resulted in extensive and abnormal elaboration of membrane protrusions and an increase in axon branching, highlighting the importance of the ERM-actin interaction in axon development. Together, our data indicate that L1 binds directly to members of the ERM family and suggest this association may coordinate aspects of axonal morphogenesis.


Assuntos
Moléculas de Adesão Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Glicoproteínas de Membrana/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Fatores de Transcrição/metabolismo , Actinas/metabolismo , Complexo 2 de Proteínas Adaptadoras , Proteínas Adaptadoras de Transporte Vesicular , Substituição de Aminoácidos , Animais , Anquirinas/metabolismo , Antígenos de Superfície/química , Antígenos de Superfície/genética , Antígenos de Superfície/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Diferenciação Celular , Citoplasma/química , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Hipocampo/citologia , Humanos , Complexo Antígeno L1 Leucocitário , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Morfogênese , Moléculas de Adesão de Célula Nervosa/química , Moléculas de Adesão de Célula Nervosa/genética , Células PC12 , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição/química , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA