Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Cell Physiol ; 229(4): 471-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24105809

RESUMO

Poly-N-acetyllactosamine (PLN) is a unique glycan composed of repeating units of the common disaccharide (Galß1,4-GlcNAcß1,3)n . The expression of PLN on glycoprotein core structures minimally requires enzyme activities for ß1,4-galactosyltransferase (ß4GalT) and ß1,3-N-acetylglucosminyltransferase (ß3GnT). Because ß4GalTs are ubiquitous in most cells, PLN expression is generally ascribed to the tissue-specific transcription of eight known ß3GnT genes in mice. In the olfactory epithelium (OE), ß3GnT2 regulates expression of extended PLN chains that are essential for axon guidance and neuronal survival. N-glycan branching and core composition, however, can also modulate the extent of PLN modification. Here, we show for the first time that the ß1,6-branching glycosyltransferase GCNT2 (formerly known as IGnT) is expressed at high levels specifically in the OE and other sensory ganglia. Postnatally, GCNT2 is maintained in mature olfactory neurons that co-express ß3GnT2 and PLN. This highly specific co-expression suggests that GCNT2 and ß3GnT2 function cooperatively in PLN synthesis. In support of this, ß3GnT2 and GCNT2 co-transfection in HEK293T cells results in high levels of PLN expression on the cell surface and on adenylyl cyclase 3, a major carrier of PLN glycans in the OE. These data clearly suggest that GCNT2 functions in vivo together with ß3GnT2 to determine PLN levels in olfactory neurons by regulating ß1,6-branches that promote PLN extension.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , N-Acetilglucosaminiltransferases/metabolismo , N-Acetilexosaminiltransferases/metabolismo , Polissacarídeos/biossíntese , Animais , Embrião de Mamíferos/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , N-Acetilglucosaminiltransferases/genética , N-Acetilexosaminiltransferases/genética , Gravidez
2.
Mol Cell Neurosci ; 52: 73-86, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23006775

RESUMO

Vomeronasal sensory neurons (VSNs) extend axons to the accessory olfactory bulb (AOB) where they form synaptic connections that relay pheromone signals to the brain. The projections of apical and basal VSNs segregate in the AOB into anterior (aAOB) and posterior (pAOB) compartments. Although some aspects of this organization exhibit fundamental similarities with the main olfactory system, the mechanisms that regulate mammalian vomeronasal targeting are not as well understood. In the olfactory epithelium (OE), the glycosyltransferase ß3GnT2 maintains expression of axon guidance cues required for proper glomerular positioning and neuronal survival. We show here that ß3GnT2 also regulates guidance and adhesion molecule expression in the vomeronasal system in ways that are partially distinct from the OE. In wildtype mice, ephrinA5(+) axons project to stereotypic subdomains in both the aAOB and pAOB compartments. This pattern is dramatically altered in ß3GnT2(-/-) mice, where ephrinA5 is upregulated exclusively on aAOB axons. Despite this, apical and basal VSN projections remain strictly segregated in the null AOB, although some V2r1b axons that normally project to the pAOB inappropriately innervate the anterior compartment. These fibers appear to arise from ectopic expression of V2r1b receptors in a subset of apical VSNs. The homotypic adhesion molecules Kirrel2 and OCAM that facilitate axon segregation and glomerular compartmentalization in the main olfactory bulb are ablated in the ß3GnT2(-/-) aAOB. This loss is accompanied by a two-fold increase in the total number of V2r1b glomeruli and a failure to form morphologically distinct glomeruli in the anterior compartment. These results identify a novel function for ß3GnT2 glycosylation in maintaining expression of layer-specific vomeronasal receptors, as well as adhesion molecules required for proper AOB glomerular formation.


Assuntos
Axônios/metabolismo , Moléculas de Adesão Celular/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Bulbo Olfatório/metabolismo , Órgão Vomeronasal/inervação , Animais , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Knockout , Órgão Vomeronasal/metabolismo
3.
J Neurosci ; 31(17): 6576-86, 2011 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-21525298

RESUMO

In the olfactory epithelium (OE), odorant receptor stimulation generates cAMP signals that function in both odor detection and the regulation of axon guidance molecule expression. The enzyme that synthesizes cAMP, adenylyl cyclase 3 (AC3), is coexpressed in olfactory sensory neurons (OSNs) with poly-N-acetyllactosamine (PLN) oligosaccharides determined by the glycosyltransferase ß3GnT2. The loss of either enzyme results in similar defects in olfactory bulb (OB) innervation and OSN survival, suggesting that glycosylation may be important for AC3 function. We show here that AC3 is extensively modified with N-linked PLN, which is essential for AC3 activity and localization. On Western blots, AC3 from the wild-type OE migrates diffusely as a heavily glycosylated 200 kDa band that interacts with the PLN-binding lectin LEA. AC3 from the ß3GnT2(-/-) OE loses these PLN modifications, migrating instead as a 140 kDa glycoprotein. Furthermore, basal and forskolin-stimulated cAMP production is reduced 80-90% in the ß3GnT2(-/-) OE. Although AC3 traffics normally to null OSN cilia, it is absent from axon projections that aberrantly target the OB. The cAMP-dependent guidance receptor neuropilin-1 is also lost from ß3GnT2(-/-) OSNs and axons, while semaphorin-3A ligand expression is upregulated. In addition, kirrel2, a mosaically expressed adhesion molecule that functions in axon sorting, is absent from ß3GnT2(-/-) OB projections. These results demonstrate that PLN glycans are essential in OSNs for proper AC3 localization and function. We propose that the loss of cAMP-dependent guidance cues is also a critical factor in the severe axon guidance defects observed in ß3GnT2(-/-) mice.


Assuntos
Adenilil Ciclases/metabolismo , Axônios/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , N-Acetilglucosaminiltransferases/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Mucosa Olfatória , Transdução de Sinais/fisiologia , Adenilil Ciclases/genética , Fatores Etários , Amino Açúcares/metabolismo , Animais , Animais Recém-Nascidos , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , AMP Cíclico/metabolismo , Proteína GAP-43/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Lectinas/metabolismo , Camundongos , Camundongos Knockout , N-Acetilglucosaminiltransferases/deficiência , Proteínas do Tecido Nervoso , Neuropilina-1/genética , Neuropilina-1/metabolismo , Mucosa Olfatória/citologia , Mucosa Olfatória/crescimento & desenvolvimento , Mucosa Olfatória/metabolismo , Receptores de Superfície Celular/metabolismo
4.
J Cell Biochem ; 112(10): 2663-71, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21618591

RESUMO

The olfactory system presents a practical model for investigating basic mechanisms involved in patterning connections between peripheral sensory neurons and central targets. Our understanding of olfactory map formation was advanced greatly by the discovery of cAMP signaling as an important determinant of glomerular positioning in the olfactory bulb. Additionally, several cell adhesion molecules have been identified recently that are proposed to regulate homotypic interactions among projecting axons. From these studies a model has emerged to partially explain the wiring of axons from widely dispersed neuron populations in the nasal cavity to relatively stereotyped glomerular positions. These advances have revitalized interest in axon guidance molecules in establishing olfactory topography, but also open new questions regarding how these patterns of guidance cues are established and function, and what other pathways, such as glycosylation, might be involved. This review summarizes the current state of this field and the important molecules that impact on cAMP-dependent mechanism in olfactory axon guidance.


Assuntos
Axônios/metabolismo , Moléculas de Adesão Celular/metabolismo , Bulbo Olfatório/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Animais , Humanos , Bulbo Olfatório/citologia , Condutos Olfatórios/fisiologia , Transdução de Sinais/fisiologia
5.
Endocrinology ; 149(9): 4596-604, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18499748

RESUMO

Hypothalamic GnRH neurons are essential for initiation and regulation of reproductive function. In addition to pituitary gonadotrope stimulation, activity of GnRH through its receptor (GnRHR) has been suggested to include autocrine regulation of the GnRH neuron. Two hypogonadal mouse strains, the Gnrh1 mutant (hpg) mice and Gnrhr mutant mice were used to investigate the potential role of GnRH signaling in the proper development and maintenance of GnRH neurons. Immunocytochemical analysis of heterozygous hpg mice revealed a GnRH neuron population that was normal in size and distribution, indicating no effect from reduced Gnrh1 gene dosage on the neurons themselves. To visualize GnRH neurons in homozygous GnRH-deficient hpg mice, heterozygous hpg mice were crossed with GnRH-green fluorescent protein (GFP) transgenic mice with targeted expression of the GFP reporter gene in GnRH neurons. Analysis of forebrains of homozygous hpg/GFP-positive mice immunostained for GFP revealed a normal population size and appropriate distribution of GnRH neurons in hpg mice, with immunoreactive neuronal processes present at the median eminence. Similarly, adult mice deficient in functional GnRHR possessed a full complement of GnRH neurons in the basal forebrain that was indistinguishable from the distribution of GnRH neurons in their wild-type counterparts. Moreover, hpg/GFP neurons retained the ability to generate spontaneous bursts of action potential firing activity, suggesting that GnRH peptide is not required for this function. These data establish that autocrine-paracrine GnRH-signaling is not a prerequisite for the developmental migration of GnRH neurons into the brain or for the projection of GnRH neurosecretory axons.


Assuntos
Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Hipogonadismo/patologia , Neurônios/patologia , Receptores LHRH/genética , Animais , Comunicação Autócrina/genética , Contagem de Células , Movimento Celular/genética , Tamanho Celular , Eletrofisiologia , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipogonadismo/genética , Hipogonadismo/metabolismo , Hipogonadismo/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Modelos Biológicos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neurônios/metabolismo , Neurônios/fisiologia , Comunicação Parácrina/genética , Receptores LHRH/metabolismo
6.
J Neurosci ; 26(25): 6834-40, 2006 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-16793890

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons migrate from the vomeronasal organ (VNO) in the nasal compartment to the basal forebrain in mice, beginning on embryonic day 11 (E11). These neurons use vomeronasal axons as guides to migrate through the nasal mesenchyme. Most GnRH neurons then migrate along the caudal branch of the vomeronasal nerve to reach the hypothalamus. We show here that stromal cell-derived factor-1 [SDF-1, also known as chemokine C-X-C motif ligand 12 (CXCL12)] is expressed in the embryonic nasal mesenchyme from as early as E10 in an increasing rostral to caudal gradient that is most intense at the border of the nasal mesenchyme and the telencephalon. Chemokine C-X-C motif receptor 4 (CXCR4), the receptor for SDF-1, is expressed by neurons in the olfactory epithelium and VNO. Cells derived from these sensory epithelia, including migrating GnRH neurons and ensheathing glial precursors of the migrating mass (MM), also express CXCR4, suggesting that they may use SDF-1 as a chemokine. In support of this, most GnRH neurons of CXCR4-/- mice fail to exit the VNO at E13, and comparatively few GnRH neurons reach the forebrain. There is also a significant decrease in the total number of GnRH neurons in CXCR4-/- mice and an increase in cell death within the VNO relative to controls. The MM is smaller in CXCR4-/- mice, suggesting that some MM cells also require SDF-1/CXCR4 function for migration and survival.


Assuntos
Movimento Celular/fisiologia , Quimiocinas CXC/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/fisiologia , Prosencéfalo/citologia , Receptores CXCR4/fisiologia , Animais , Morte Celular/fisiologia , Quimiocina CXCL12 , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Imuno-Histoquímica/métodos , Hibridização In Situ/métodos , Marcação In Situ das Extremidades Cortadas/métodos , Camundongos , Camundongos Knockout , Receptores CXCR4/deficiência , Tubulina (Proteína)/metabolismo , Órgão Vomeronasal/citologia
7.
J Mol Histol ; 38(6): 543-53, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17605079

RESUMO

Despite the relatively simplified organization of the olfactory epithelium (OE), our understanding of the factors that regulate its cellular diversity is limited. Genetic and localization studies suggest that Notch signaling may be important in this process. We characterize here a population of Notch1 (+) olfactory basal cells in embryonic mice that coordinately express both the Notch effector Hes5 and the glycosyltransferase Lfng. These cells are distinct from Mash1(+) neuronal precursors, but give rise to sensory neurons, suggesting that Notch1 signals may in part function to maintain a neurogenic progenitor pool. Furthermore, Lfng(+) cells also generate a population of cells in the migratory mass that appear to be ensheathing glial precursors, indicating potential multipotency in these progenitors. The Notch ligand Dll4 is expressed by basal OE cells that are interspersed with Notch1(+) progenitors during later OE neurogenesis. In contrast, mice deficient in Dll1 exhibit a smaller OE and a loss of Hes5 expression, indicating an earlier function in olfactory progenitor cell development. Taken together, these results further support a role for Notch signaling in the regulation of olfactory neurogenesis and cell diversity.


Assuntos
Mucosa Olfatória/metabolismo , Receptor Notch1/metabolismo , Células-Tronco/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação ao Cálcio , Expressão Gênica , Glicosiltransferases/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ligantes , Proteínas de Membrana/metabolismo , Camundongos , Mucosa Olfatória/citologia , Mucosa Olfatória/embriologia , Proteínas Repressoras/metabolismo , Células-Tronco/citologia
8.
Semin Reprod Med ; 25(5): 305-12, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17710726

RESUMO

Neurons that synthesize and secrete the decapeptide gonadotropin-releasing hormone-1 (GnRH-1) to control the reproductive axis originate in the olfactory placode/vomeronasal organ of the olfactory system of mammals and migrate along vomeronasal nerves to the cribriform plate, which marks the boundary between the peripheral olfactory system and the forebrain. Migrating GnRH-1 neurons follow a branch of the vomeronasal nerve caudally into the hypothalamus, where they extend processes to the median eminence and halt their migration. The release of GnRH-1 into the capillaries of the median eminence starts the cascade that activates pituitary gonadotropin (luteinizing hormone and follicle-stimulating hormone) production and secretion. Failure of these neurons to complete their migration results in failure of the reproductive axis. In some cases, failed migration is linked to the loss of the sense of smell (anosmia). The mechanisms that regulate migration of GnRH-1 neurons along this complex pathway are incompletely understood. Recent studies have revealed an important role for a series of strategically located soluble factors that regulate different aspects of GnRH-1 neuron migration at specific locations along their migratory route. This review focuses on the different mechanisms used by these factors to regulate migration of GnRH-1 neurons.


Assuntos
Movimento Celular , Hormônio Liberador de Gonadotropina/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Neurônios/metabolismo , Precursores de Proteínas/metabolismo , Reprodução , Transdução de Sinais , Animais , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Humanos , Hipogonadismo/metabolismo , Hipogonadismo/fisiopatologia , Sistema Hipotálamo-Hipofisário/crescimento & desenvolvimento , Síndrome de Kallmann/metabolismo , Síndrome de Kallmann/fisiopatologia , Kisspeptinas , Fatores de Crescimento Neural/metabolismo , Netrina-1 , Condutos Olfatórios/crescimento & desenvolvimento , Condutos Olfatórios/metabolismo , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/metabolismo , Receptores CXCR4/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1 , Proteínas Supressoras de Tumor/metabolismo
9.
J Neurosci ; 25(8): 1894-903, 2005 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-15728829

RESUMO

During embryonic development, axons from sensory neurons in the olfactory epithelium (OE) extend into the olfactory bulb (OB) where they synapse with projection neurons and form glomerular structures. To determine whether glycans play a role in these processes, we analyzed mice deficient for the glycosyltransferase beta1,3-N-acetylglucosaminyltransferase 1 (beta3GnT1), a key enzyme in lactosamine glycan synthesis. Terminal lactosamine expression, as shown by immunoreactivity with the monoclonal antibody 1B2, is dramatically reduced in the neonatal null OE. Postnatal beta3GnT1-/- mice exhibit severely disorganized OB innervation and defective glomerular formation. Beginning in embryonic development, specific subsets of odorant receptor-expressing neurons are progressively lost from the OE of null mice, which exhibit a postnatal smell perception deficit. Axon guidance errors and increased neuronal cell death result in an absence of P2, I7, and M72 glomeruli, indicating a reduction in the repertoire of odorant receptor-specific glomeruli. By approximately 2 weeks of age, lactosamine is unexpectedly reexpressed in sensory neurons of null mice through a secondary pathway, which is accompanied by the regrowth of axons into the OB glomerular layer and the return of smell perception. Thus, both neonatal OE degeneration and the postnatal regeneration are lactosamine dependent. Lactosamine expression in beta3GnT1-/- mice is also reduced in pheromone-receptive vomeronasal neurons and dorsal root ganglion cells, suggesting that beta3GnT1 may perform a conserved function in multiple sensory systems. These results reveal an essential role for lactosamine in sensory axon pathfinding and in the formation of OB synaptic connections.


Assuntos
Amino Açúcares/fisiologia , Axônios/fisiologia , N-Acetilglucosaminiltransferases/fisiologia , Neurônios Aferentes/fisiologia , Bulbo Olfatório/citologia , Processamento de Proteína Pós-Traducional , Amino Açúcares/biossíntese , Animais , Sequência de Carboidratos , Morte Celular , Indução Enzimática , Comportamento Alimentar , Gânglios Espinais/citologia , Glicosilação , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , N-Acetilglucosaminiltransferases/deficiência , N-Acetilglucosaminiltransferases/genética , Degeneração Neural , Regeneração Nervosa , Neurônios Aferentes/ultraestrutura , Bulbo Olfatório/enzimologia , Bulbo Olfatório/patologia , Polissacarídeos/biossíntese , Polissacarídeos/fisiologia , RNA Mensageiro/biossíntese , Receptores Odorantes/fisiologia , Sinapses/fisiologia , Órgão Vomeronasal/citologia
10.
Endocrinology ; 147(3): 1159-65, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16373413

RESUMO

Neurons that synthesize GnRH are critical brain regulators of the reproductive axis, yet they originate outside the brain and must migrate over long distances and varied environments to get to their appropriate positions during development. Many studies, past and present, are providing clues for the types of molecules encountered and movements expected along the migratory route. Recent studies provide real-time views of the behavior of GnRH neurons in the context of in vitro preparations that model those in vivo. Live images provide direct evidence of the changing behavior of GnRH neurons in their different environments, showing that GnRH neurons move with greater frequency and with more alterations in direction after they enter the brain. The heterogeneity of molecular phenotypes for GnRH neurons likely ensures that multiple external factors will be found that regulate the migration of different portions of the GnRH neuronal population at different steps along the route. Molecules distributed in gradients both in the peripheral olfactory system and basal forebrain may be particularly influential in directing the appropriate movement of GnRH neurons along their arduous migration. Molecules that mediate the adhesion of GnRH neurons to changing surfaces may also play critical roles. It is likely that the multiple external factors converge on selective signal transduction pathways to engage the mechanical mechanisms needed to modulate GnRH neuronal movement and ultimately migration.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Vias Neurais , Neurônios/metabolismo , Animais , Encéfalo/metabolismo , Movimento Celular , Citoesqueleto/metabolismo , Humanos , Camundongos , Modelos Biológicos , Bulbo Olfatório/metabolismo , Condutos Olfatórios , Fenótipo , Prosencéfalo/metabolismo , Transdução de Sinais
11.
Endocrinology ; 146(1): 463-8, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15486219

RESUMO

Neurons that synthesize GnRH control the reproductive axis and migrate over long distances and through different environments during development. Prior studies provided strong clues for the types of molecules encountered and movements expected along the migratory route. However, our studies provide the first real-time views of the behavior of GnRH neurons in the context of an in vitro preparation that maintains conditions comparable to those in vivo. The live views provide direct evidence of the changing behavior of GnRH neurons in their different environments, showing that GnRH neurons move with greater frequency and with more changes in direction after they enter the brain. Perturbations of guiding fibers distal to moving GnRH neurons in the nasal compartment influenced movement without detectable changes in the fibers in the immediate vicinity of moving GnRH neurons. This suggests that the use of fibers by GnRH neurons for guidance may entail selective signaling in addition to mechanical guidance. These studies establish a model to evaluate the influences of specific molecules that are important for their migration.


Assuntos
Sistemas Computacionais , Hormônio Liberador de Gonadotropina/metabolismo , Microscopia de Vídeo , Neurônios/fisiologia , Animais , Bicuculina/farmacologia , Encéfalo/embriologia , Movimento Celular , Forma Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/inervação , Antagonistas GABAérgicos/farmacologia , Antagonistas de Receptores de GABA-A , Proteínas de Fluorescência Verde , Imuno-Histoquímica , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Vias Neurais/embriologia , Neurônios/citologia , Neurônios/metabolismo , Nariz/embriologia , Fixação de Tecidos
12.
Endocrinology ; 144(6): 2566-79, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12746320

RESUMO

gamma-Aminobutyric acid (GABA) inhibits the embryonic migration of GnRH neurons and regulates hypothalamic GnRH release. A subset of GnRH neurons expresses GABA along their migratory route in the nasal compartment before entering the brain, suggesting that GABA produced by GnRH neurons may help regulate the migratory process. To examine this hypothesis and the possibility that persistence of GABA production by GnRH neurons may affect subsequent reproductive function, we generated transgenic mice in which the expression of glutamic acid decarboxylase-67 (GAD-67), a key enzyme in GABA synthesis, is targeted to GnRH neurons under the control of the GnRH gene promoter. On embryonic d 15, when GnRH neurons are still migrating, the transgenic animals had more GnRH neurons in aberrant locations in the cerebral cortex and fewer neurons reaching the hypothalamic-preoptic region, whereas migration into the brain was not affected. Hypothalamic GnRH content in mutant mice was low during the first week of postnatal life, increasing to normal values during infantile development (second week after birth) in the presence of increased pulsatile GnRH release. Consistent with these changes, serum LH and FSH levels were also elevated. Gonadotropin release returned to normal values by the time steroid negative feedback became established (fourth week of life). Ovariectomy at this time demonstrated an enhanced gonadotropin response in transgenic animals. Although the onset of puberty, as assessed by the age at vaginal opening and first ovulation, was not affected in the mutant mice, estrous cyclicity and adult reproductive capacity were disrupted. Mutant mice had reduced litter sizes, increased time intervals between deliveries of litters, and a shorter reproductive life span. Thus, GABA produced within GnRH neurons does not delay GnRH neuronal migration, but instead serves as a developmental cue that increases the positional diversity of these neurons within the basal forebrain. In addition, the results suggest that the timely termination of GABA production within the GnRH neuronal network is a prerequisite for normal reproductive function. The possibility arises that similar abnormalities in GABA homeostasis may contribute to syndromes of hypothalamic amenorrhea/oligomenorrhea in humans.


Assuntos
Movimento Celular/fisiologia , Glutamato Descarboxilase/genética , Hormônio Liberador de Gonadotropina/metabolismo , Isoenzimas/genética , Neurônios/citologia , Reprodução/fisiologia , Animais , Animais Recém-Nascidos , Ciclo Estral/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Glutamato Descarboxilase/metabolismo , Hipotálamo/citologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/fisiologia , Isoenzimas/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/enzimologia , Regiões Promotoras Genéticas/genética , Fluxo Pulsátil , Ratos , Ácido gama-Aminobutírico/biossíntese , Ácido gama-Aminobutírico/metabolismo
13.
Artigo em Inglês | MEDLINE | ID: mdl-24130552

RESUMO

The hypothalamic-pituitary-gonadal axis (HPG) is a complex neuroendocrine circuit involving multiple levels of regulation. Kisspeptin neurons play essential roles in controlling the HPG axis from the perspectives of puberty onset, oscillations of gonadotropin releasing hormone (GnRH) neuron activity, and the pre-ovulatory LH surge. The current studies focus on the expression of kisspeptin during murine fetal development using in situ hybridization (ISH), quantitative reverse transcription real-time PCR (QPCR), and immunocytochemistry. Expression of mRNA coding for kisspeptin (KISS1) and its receptor KISS1R was observed at embryonic (E) day 13 by ISH. At E13 and other later ages examined, Kiss1 signal in individual cells within the arcuate nucleus (ARC) appeared stronger in females than males. ISH examination of agonadal steroidogenic factor-1 (Sf1) knockout mice revealed that E17 XY knockouts (KO) resembled wild-type (WT) XX females. These findings raise the possibility that gonadal hormones modulate the expression of Kiss1 in the ARC prior to birth. The sex and genotype differences were tested quantitatively by QPCR experiments in dissected hypothalami from mice at E17 and adulthood. Females had significantly more Kiss1 than males at both ages, even though the number of cells detected by ISH was similar. In addition, QPCR revealed a significant difference in the amount of Kiss1 mRNA in Sf1 mice with WT XY mice expressing less than XY KO and XX mice of both genotypes. The detection of immunoreactive KISS1 in perikarya of the ARC at E17 indicates that early mRNA is translated to peptide. The functional significance of this early expression of Kiss1 awaits elucidation.

14.
Neural Dev ; 7: 17, 2012 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-22559903

RESUMO

BACKGROUND: The defining feature of the main olfactory system in mice is that each olfactory sensory neuron expresses only one of more than a thousand different odorant receptor genes. Axons expressing the same odorant receptor converge onto a small number of targets in the olfactory bulb such that each glomerulus is made up of axon terminals expressing just one odorant receptor. It is thought that this precision in axon targeting is required to maintain highly refined odor discrimination. We previously showed that ß3GnT2(-/-) mice have severe developmental and axon guidance defects. The phenotype of these mice is similar to adenylyl cyclase 3 (AC3) knockout mice largely due to the significant down-regulation of AC3 activity in ß3GnT2(-/-) neurons. RESULTS: Microarray analysis reveals that nearly one quarter of all odorant receptor genes are down regulated in ß3GnT2(-/-) mice compared to controls. Analysis of OR expression by quantitative PCR and in situ hybridization demonstrates that the number of neurons expressing some odorant receptors, such as mOR256-17, is increased by nearly 60% whereas for others such as mOR28 the number of neurons is decreased by more than 75% in ß3GnT2(-/-) olfactory epithelia. Analysis of axon trajectories confirms that many axons track to inappropriate targets in ß3GnT2(-/-) mice, and some glomeruli are populated by axons expressing more than one odorant receptor. Results show that mutant mice perform nearly as well as control mice in an odor discrimination task. In addition, in situ hybridization studies indicate that the expression of several activity dependent genes is unaffected in ß3GnT2(-/-) olfactory neurons. CONCLUSIONS: Results presented here show that many odorant receptors are under-expressed in ß3GnT2(-/-) mice and further demonstrate that additional axon subsets grow into inappropriate targets or minimally innervate glomeruli in the olfactory bulb. Odor evoked gene expression is unchanged and ß3GnT2(-/-) mice exhibit a relatively small deficit in their ability to discriminate divergent odors. Results suggest that despite the fact that ß3GnT2(-/-) mice have decreased AC3 activity, decreased expression of many ORs, and display many axon growth and guidance errors, odor-evoked activity in cilia of mutant olfactory neurons remains largely intact.


Assuntos
Axônios/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Percepção Olfatória , Neurônios Receptores Olfatórios/metabolismo , Receptores Odorantes/metabolismo , Animais , Animais Recém-Nascidos , Discriminação Psicológica , Regulação para Baixo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , N-Acetilglucosaminiltransferases/deficiência , Bulbo Olfatório/metabolismo , Condutos Olfatórios/metabolismo , Receptores Odorantes/genética , Transdução de Sinais/fisiologia
15.
J Neurosci Res ; 86(1): 11-7, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17549746

RESUMO

The olfactory system represents a complex model for the investigation of factors that influence the guidance of sensory axon populations to specific targets in the CNS. In the mouse, the projections of approximately 1,000 neuronal subsets, each defined by expression of a distinct odorant receptor (OR), converge at unique glomerular loci in the olfactory bulb (OB). Unlike the case in other sensory systems, proper guidance is achieved without benefit of any known cues in the target itself that are capable of attracting or repelling specific axons. It has long been argued that OR proteins are the critical molecules orchestrating guidance. However, recent studies suggest that axon identity may be dependent on the graded expression of a variety of unique olfactory axon guidance cues. This review focuses attention on these non-OR factors and their roles in olfactory axon guidance.


Assuntos
Axônios/fisiologia , Condutos Olfatórios/citologia , Condutos Olfatórios/fisiologia , Animais , Modelos Biológicos , Receptores Odorantes/fisiologia , Transdução de Sinais/fisiologia
16.
Dev Neurobiol ; 67(12): 1627-40, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17567839

RESUMO

During embryonic development, olfactory sensory neurons extend axons that form synapses with the dendrites of projection neurons in glomeruli of the olfactory bulb (OB). The glycosyltransferase beta3GnT1 regulates the expression of 1B2-reactive lactosamine glycans that are mosaically distributed among glomeruli. In newborn beta3GnT1-/- mice, lactosamine expression is lost, and many glomeruli fail to form. To determine the role of lactosamine in OB targeting, we analyzed the trajectories of specific OR axon populations and their reactivity with 1B2 in beta3GnT1-/- mice. mI7 axons and P2 axons, both of which are weakly 1B2+ in wild-type mice, fail to grow to their normal positions in the glomerular layer during early postnatal development and never recover in adult mutant mice. In contrast, many M72 axons, which are always lactosamine negative in wild-type mice, survive but are misguided to the extreme anterior OB in neonatal mutant mice and persist as heterotypic glomeruli, even in adult null mice. These results show that the loss of lactosamine differentially affects each OR population. Those that lose their normal expression of lactosamine fail to form stable connections with mitral and tufted cells in the OB, disappear during early postnatal development, and do not recover in adults. Neurons that are normally lactosamine negative, survive early postnatal degeneration in beta3GnT1-/- mice but extend axons that converge on inappropriate targets in the mutant OB.


Assuntos
Amino Açúcares/biossíntese , Neurônios Aferentes/citologia , Bulbo Olfatório/citologia , Condutos Olfatórios/citologia , Animais , Animais Recém-Nascidos , Axônios/metabolismo , Axônios/ultraestrutura , Glicosiltransferases/deficiência , Imuno-Histoquímica , Camundongos , Camundongos Mutantes , Neurônios Aferentes/metabolismo , Bulbo Olfatório/crescimento & desenvolvimento , Bulbo Olfatório/metabolismo , Condutos Olfatórios/crescimento & desenvolvimento , Condutos Olfatórios/metabolismo
17.
J Cell Physiol ; 210(2): 290-7, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17111357

RESUMO

The olfactory system is a remarkable model for investigating the factors that influence the guidance of sensory axon populations to specific targets in the CNS. Since the initial discovery of the vast odorant receptor (ORs) gene family in rodents and the subsequent finding that these molecules directly influence targeting, several additional olfactory axon guidance cues have been identified. Two of these, ephrins and semaphorins, have well-established functions in patterning axon connections in other systems. In addition, lactosamine-containing glycans are also required for proper targeting and maintenance of olfactory axons, and may also function in other sensory regions. It is now apparent that these and likely other additional molecules are required along with ORs to orchestrate the complex pattern of convergence and divergence that is unique to the olfactory system.


Assuntos
Cones de Crescimento/metabolismo , Fatores de Crescimento Neural/metabolismo , Condutos Olfatórios/embriologia , Condutos Olfatórios/metabolismo , Neurônios Receptores Olfatórios/embriologia , Neurônios Receptores Olfatórios/metabolismo , Animais , Diferenciação Celular/fisiologia , Cones de Crescimento/ultraestrutura , Humanos , Regeneração Nervosa/fisiologia , Neuroglia/citologia , Condutos Olfatórios/citologia , Neurônios Receptores Olfatórios/citologia
18.
Eur J Neurosci ; 24(3): 654-60, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16930397

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons are derived from progenitor cells in the olfactory placodes and migrate from the vomeronasal organ (VNO) across the cribriform plate into the forebrain. At embryonic day (E)12 in the mouse most of these neurons are still in the nasal compartment but by E15 most GnRH neurons have migrated into the forebrain. Glycoconjugates with carbohydrate chains containing terminal lactosamine are expressed by neurons in the main olfactory epithelium and in the VNO. One of the key enzymes required to regulate the synthesis and expression of lactosamine, beta1,3-N-acetylglucosaminyltransferase-1 (beta3GnT1), is strongly expressed by neurons in the olfactory epithelium and VNO, and on neurons migrating out of the VNO along the GnRH migratory pathway. Immunocytochemical analysis of lactosamine and GnRH in embryonic mice reveals that the percentage of lactosamine+-GnRH+ double-labeled neurons decreases from > 80% at E13, when migration is near its peak, to approximately 30% at E18.5, when most neurons have stopped migrating. In beta3GnT1-/- mice, there is a partial loss of lactosamine expression on GnRH neurons. Additionally, a greater number of GnRH neurons were retained in the nasal compartment of null mice at E15 while fewer GnRH neurons were detected later in embryonic development in the ventral forebrain. These results suggest that the loss of lactosamine on a subset of GnRH neurons impeded the rate of migration from the nose to the brain.


Assuntos
Amino Açúcares/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Mucosa Olfatória/embriologia , Prosencéfalo/embriologia , Órgão Vomeronasal/embriologia , Animais , Contagem de Células , Diferenciação Celular/genética , Movimento Celular/genética , Regulação para Baixo/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , N-Acetilglucosaminiltransferases/genética , Bulbo Olfatório/citologia , Bulbo Olfatório/embriologia , Bulbo Olfatório/metabolismo , Mucosa Olfatória/citologia , Mucosa Olfatória/metabolismo , Condutos Olfatórios/citologia , Condutos Olfatórios/embriologia , Condutos Olfatórios/metabolismo , Prosencéfalo/citologia , Prosencéfalo/metabolismo , Órgão Vomeronasal/citologia , Órgão Vomeronasal/metabolismo
19.
Eur J Neurosci ; 19(1): 11-20, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14750959

RESUMO

Luteinizing hormone-releasing hormone (LHRH) neurons migrate from the vomeronasal organ (VNO) to the forebrain in all mammals studied. In mice, the direction of LHRH neuron migration is dependent upon axons that originate in the VNO, but bypass the olfactory bulb and project caudally into the basal forebrain. Thus, factors that guide this unique subset of vomeronasal axons that comprise the caudal vomeronasal nerve (cVNN) are candidates for regulating the migration of LHRH neurons. We previously showed that deleted in colorectal cancer (DCC) is expressed by neurons that migrate out of the VNO during development [Schwarting et al. (2001) J. Neurosci., 21, 911-919]. We examined LHRH neuron migration in Dcc-/- mice and found that trajectories of the cVNN and positions of LHRH neurons are abnormal. Here we extend these studies to show that cVNN trajectories and LHRH cell migration in netrin 1 (Ntn1) mutant mice are also abnormal. Substantially reduced numbers of LHRH neurons are found in the basal forebrain and many LHRH neurons migrate into the cerebral cortex of Ntn1 knockout mice. In contrast, migration of LHRH cells is normal in Unc5h3rcm mutant mice. These results are consistent with the idea that the chemoattraction of DCC+ vomeronasal axons by a gradient of netrin 1 protein in the ventral forebrain guides the cVNN, which, in turn, determines the direction of LHRH neuron migration in the forebrain. Loss of function through a genetic deletion in either Dcc or Ntn1 results in the migration of many LHRH neurons to inappropriate destinations.


Assuntos
Fatores Quimiotáticos/metabolismo , Quimiotaxia/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Fatores de Crescimento Neural/metabolismo , Vias Neurais/embriologia , Neurônios/metabolismo , Animais , Moléculas de Adesão Celular/biossíntese , Moléculas de Adesão Celular/deficiência , Moléculas de Adesão Celular/genética , Fatores Quimiotáticos/deficiência , Fatores Quimiotáticos/genética , Quimiotaxia/genética , Coristoma/genética , Coristoma/metabolismo , Receptor DCC , Feto , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Fatores de Crescimento Neural/deficiência , Fatores de Crescimento Neural/genética , Receptores de Netrina , Netrina-1 , Vias Neurais/citologia , Vias Neurais/metabolismo , Neurônios/citologia , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Telencéfalo/citologia , Telencéfalo/embriologia , Telencéfalo/metabolismo , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Órgão Vomeronasal/citologia , Órgão Vomeronasal/embriologia , Órgão Vomeronasal/metabolismo
20.
Eur J Neurosci ; 19(7): 1800-10, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15078553

RESUMO

Semaphorins are known to play an important role in axon guidance of vertebrate olfactory sensory neurons to their targets in specific glomeruli of the olfactory bulb (OB). However, it is not clear how semaphorin-mediated guidance contributes to a systematic hierarchy of cues that govern the organization of this system. Because of the putative role that odorant receptor molecules such as P2 could play in establishing appropriate glomerular destinations for growing olfactory axons, we have also determined the spatial organization of P2 glomeruli in semaphorin 3A (Sema3A) mutant mice. First, in the postnatal OB of control and Sema3A(-/-) mice, we analysed the trajectories of olfactory axons that express the Sema3A receptor, neuropilin-1 (npn-1) and the positions of npn-1(+) glomeruli. Sema3A at the ventral OB midline guides npn-1(+) axons to targets in the lateral and medial OB. Absence of Sema3A permits many npn-1 axons to terminate aberrantly in the rostral and ventral OB. Second, in Sema3A(-/-) mice, many P2 axons are abnormally distributed throughout the ventral OB nerve layer and converge in atypical locations compared with littermate controls where P2 axons converge on stereotypically located lateral and medial glomeruli. In addition to their radically altered spatial distribution, P2 glomeruli in Sema3A(-/-) mice are significantly smaller and more numerous than in heterozygote littermates. These data show that Sema3A is an important repulsive olfactory guidance cue that establishes restricted npn-1(+) subcompartments in the olfactory bulb. Furthermore, Sema3A plays a key role in the convergence of axons expressing the odorant receptor P2 onto their appropriate targets.


Assuntos
Axônios/metabolismo , Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas do Tecido Nervoso/metabolismo , Bulbo Olfatório/citologia , Neurônios Receptores Olfatórios/metabolismo , Receptores Odorantes/metabolismo , Semaforina-3A , Animais , Animais Recém-Nascidos , Proteínas de Transporte/genética , Tamanho Celular/genética , Embrião de Mamíferos , Imuno-Histoquímica/métodos , Hibridização In Situ/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Neuropilina-1/metabolismo , Bulbo Olfatório/metabolismo , Coloração e Rotulagem/métodos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa