Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Front Neuroendocrinol ; 35(1): 140-58, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24355523

RESUMO

Comorbidity of major depressive disorder (MDD) and cardiovascular disease (CVD) represents the fourth leading cause of morbidity and mortality worldwide, and women have a two times greater risk than men. Thus understanding the pathophysiology has widespread implications for attenuation and prevention of disease burden. We suggest that sex-dependent MDD-CVD comorbidity may result from alterations in fetal programming consequent to the prenatal maternal environments that produce excess glucocorticoids, which then drive sex-dependent developmental alterations of the fetal hypothalamic-pituitary-adrenal (HPA) axis circuitry impacting mood, stress regulation, autonomic nervous system (ANS), and the vasculature in adulthood. Evidence is consistent with the hypothesis that disruptions of pathways associated with gamma aminobutyric acid (GABA) in neuronal and vascular development and growth factors have critical roles in key developmental periods and adult responses to injury in heart and brain. Understanding the potential fetal origins of these sex differences will contribute to development of novel sex-dependent therapeutics.


Assuntos
Doenças Cardiovasculares/metabolismo , Depressão/metabolismo , Desenvolvimento Fetal/fisiologia , Caracteres Sexuais , Estresse Fisiológico , Animais , Doenças Cardiovasculares/fisiopatologia , Depressão/fisiopatologia , Feminino , Humanos , Masculino , Risco
2.
Pflugers Arch ; 465(5): 585-94, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23503726

RESUMO

There is a strong and growing literature showing that key aspects of brain development may be critical antecedents of adult physiology and behavior or may lead to physiological and psychiatric disorders in adulthood. Many are significantly influenced by sex-dependent factors. Neurons of the paraventricular nucleus (PVN) of the hypothalamus occupy a key position in regulating homeostatic, neuroendocrine, and behavioral functions. This brain area is a critical link for our understanding of the etiology of a number of disorders with components ranging from mood to feeding and energy balance and to autonomic nervous system regulation. Thus, based on common brain circuitry, the PVN may be a critical anatomical intersection for understanding comorbidities among depression, obesity, and cardiovascular risk. Historically, the majority of approaches to brain development examine neuronal, glial, and vascular factors independently, with notably less emphasis on vascular contributions. The realization that the PVN undergoes a unique vascular developmental process places added value on discerning the cellular and molecular mechanisms that drive its late-onset angiogenesis and further implications for neuronal differentiation and function. This has ramifications in humans for understanding chronic, and sometimes fatal, comorbidities that share sex-dependent biological bases in development through functional and anatomical intersections with the hypothalamus.


Assuntos
Doenças Cardiovasculares/fisiopatologia , Transtorno Depressivo Maior/fisiopatologia , Caracteres Sexuais , Encéfalo/metabolismo , Doenças Cardiovasculares/epidemiologia , Comorbidade , Transtorno Depressivo Maior/epidemiologia , Feminino , Humanos , Masculino
3.
Horm Metab Res ; 44(8): 619-24, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22488519

RESUMO

The paraventricular nucleus of the hypothalamus (PVN) is a cell group that plays important roles in regulating sympathetic vasomotor tone, food intake, neuroendocrine and autonomic stress responses, and cardiovascular function. The developing PVN is surrounded by neuronal elements containing, and presumably secreting, gamma-aminobutyric acid (GABA). The vasculature of the adult PVN is notably denser than in other brain regions or in the PVN during perinatal development. To characterize the postnatal angiogenic process in mice, blood vessels were analyzed at P8, 20, and 50 in rostral, mid, and caudal divisions of the PVN in males and females. Vascular changes relative to disruption of the R1 subunit of the GABA(B) receptor were evaluated at P8 and P20. For defined regions of interest within the PVN there were age dependent increases in blood vessel lengths and branching from P8 to 20 to 50 with the most notable increases in the middle region. Loss of GABA(B) receptors did not influence vascular characteristics at P8 in any region, but by P20 there was significantly (20%) less blood vessel length and branching in the mid-PVN region vs. wild type. These findings suggest that the loss of GABA(B) signaling may lead to a late developing defect in angiogenesis. The loss of vascularity with defective GABA(B) signaling suggests that neurovascular relationships in the PVN may be an important locus for understanding disorders of the hypothalamic-pituitary-adrenal axis with potential impact for psychiatric mood disorders along with other comorbid disorders that may be regulated by cells in the PVN.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/metabolismo , Núcleo Hipotalâmico Paraventricular/anatomia & histologia , Núcleo Hipotalâmico Paraventricular/irrigação sanguínea , Transdução de Sinais , Ácido gama-Aminobutírico/metabolismo , Animais , Vasos Sanguíneos/anatomia & histologia , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores de GABA-B/metabolismo
4.
Horm Metab Res ; 44(8): 577-86, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22700441

RESUMO

Hormones influence countless biological processes across an animal's lifespan. Many hormone-mediated events occur within developmental sensitive periods, during which hormones have the potential to cause permanent tissue-specific alterations in anatomy and physiology. There are numerous selective critical periods in development with different targets being affected during different periods. This review outlines the proceedings of the Hormonal Programming in Development session at the US-South American Workshop in Neuroendocrinology in August 2011. Here we discuss how gonadal steroid hormones impact various biological processes within the brain and gonads during early development and describe the changes that take place in the aging female ovary. At the cellular level, hormonal targets in the brain include neurons, glia, or vasculature. On a genomic/epigenomic level, transcription factor signaling and epigenetic changes alter the expression of critical hormone receptor genes across development and following ischemic brain insult. In addition, organizational hormone exposure alters epigenetic processes in specific brain nuclei and may be an important mediator of sexual differentiation of the neonatal brain. Brain targets of hormonal programming, such as the paraventricular nucleus of the hypothalamus, may be critical in influencing the development of peripheral targets, such as the ovary. Exposure to excess hormones can cause abnormalities in the ovary during development leading to polycystic ovarian syndrome (PCOS). Exposure to excess androgens during fetal development also has a profound effect on the development of the male reproductive system. In addition, increased activity of the sympathetic nerve and stress during early life have been linked to PCOS symptomology in adulthood. Finally, we describe how age-related decreases in fertility are linked to high levels of nerve growth factor (NGF), which enhances sympathetic nerve activity and alters ovarian function.


Assuntos
Epigênese Genética , Crescimento e Desenvolvimento/fisiologia , Hormônios/metabolismo , Envelhecimento/genética , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Crescimento e Desenvolvimento/genética , Humanos , Reprodução/genética
5.
Neuroscience ; 151(4): 1119-31, 2008 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-18248902

RESUMO

The ventromedial (VMN) and arcuate (ARC) nuclei of the hypothalamus are bilateral nuclear groups at the base of the hypothalamus that are organized through the aggregation of neurons born along the third ventricle that migrate laterally. During development, GABAergic neurons and fibers surround the forming (or primordial) VMN while neurons containing GABA receptors are found within the boundaries of the emerging nucleus. To investigate the role that GABAB receptors play in establishing the VMN, Thy-1 yellow fluorescent protein (YFP) mice were utilized for live video microscopy studies. The Thy-1 promoter drives YFP expression in regions of the hypothalamus during development. Administration of the GABAB receptor antagonist saclofen and the GABAA receptor antagonist bicuculline selectively increased the rate of VMN cell movement in slices placed in vitro at embryonic day 14, when cells that form both the ARC and VMN are migrating away from the proliferative zone surrounding the third ventricle. To further test the role of GABAB receptors in VMN development, GABAB receptor knockout mice were used to examine changes in the positions of phenotypically identified cells within the VMN. Cells containing immunoreactive estrogen receptors (ER) alpha were located in the ventrolateral quadrant of the wild type VMN. In GABABR1 knockout mice, these ERalpha positive neurons were located in more dorsal positions at postnatal day (P) 0 and P4. We conclude that GABA alters cell migration and its effect on final cell positioning may lead to changes in the circuitry and connections within specific nuclei of the developing hypothalamus.


Assuntos
Movimento Celular/fisiologia , Neurônios/fisiologia , Receptores de GABA-B/fisiologia , Núcleo Hipotalâmico Ventromedial/citologia , Animais , Animais Recém-Nascidos , Baclofeno/análogos & derivados , Baclofeno/farmacologia , Movimento Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Receptor alfa de Estrogênio/metabolismo , Antagonistas GABAérgicos/farmacologia , Técnicas In Vitro , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Receptores de GABA-A/metabolismo , Receptores de GABA-B/deficiência , Núcleo Hipotalâmico Ventromedial/embriologia , Núcleo Hipotalâmico Ventromedial/crescimento & desenvolvimento
6.
J Neurosci ; 21(3): 911-9, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11157077

RESUMO

Luteinizing hormone-releasing hormone (LHRH) neurons migrate from the vomeronasal organ (VNO) to the forebrain in all mammals studied. In mice, most LHRH neuron migration is dependent on axons that originate in the VNO but bypass the olfactory bulb and project into the basal forebrain. Thus, cues that regulate the trajectories of these vomeronasal axons are candidates for determining the destination of LHRH neurons. Using in situ hybridization techniques, we examined the expression of Deleted in colorectal cancer (DCC), a vertebrate receptor for the guidance molecule netrin-1, during development of the olfactory system. DCC is expressed by cells in the olfactory epithelium (OE) and VNO, and in cells migrating from the OE and VNO from embryonic day 11 (E11) to E14. Some DCC(+) cells on vomeronasal axons in the nose also express LHRH. However, DCC expression is downregulated beginning at E12, so few if any LHRH neurons in the forebrain also express DCC. In rat, DCC is expressed on TAG-1(+) axons that guide migrating LHRH neurons. We therefore examined LHRH neuron migration in DCC(-/-) mice and found that trajectories of the caudal vomeronasal nerve and positions of LHRH neurons are abnormal. Fewer than the normal number of LHRH neurons are found in the basal forebrain, and many LHRH neurons are displaced into the cerebral cortex of DCC(-/-) mice. These results are consistent with the idea that DCC regulates the trajectories of a subset of vomeronasal axons that guide the migration of LHRH neurons. Loss of DCC function results in the migration of many LHRH neurons to inappropriate destinations.


Assuntos
Moléculas de Adesão Celular/metabolismo , Movimento Celular/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Prosencéfalo/metabolismo , Proteínas Supressoras de Tumor , Animais , Animais Recém-Nascidos , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/farmacologia , Contagem de Células , Movimento Celular/efeitos dos fármacos , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Receptor DCC , Homozigoto , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Neurônios/citologia , Neurônios/efeitos dos fármacos , Condutos Olfatórios/citologia , Condutos Olfatórios/embriologia , Condutos Olfatórios/metabolismo , Prosencéfalo/citologia , Prosencéfalo/embriologia , RNA Mensageiro/biossíntese , Ratos , Receptores de Superfície Celular , Órgão Vomeronasal/citologia , Órgão Vomeronasal/embriologia , Órgão Vomeronasal/inervação , Órgão Vomeronasal/metabolismo
7.
Endocrinology ; 122(2): 524-30, 1988 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-3338412

RESUMO

The present studies examined the patterns of circulating testosterone (T) within 0-24 h after birth in male and female ferrets along with concomitant changes in neural aromatase activity and hypothalamic concentrations of estradiol (E2). Plasma and brain samples were obtained 0 and 2 h (cesarean delivery) or 0, 2, 12, and 24 h (natural delivery) after birth. Plasma T levels were significantly higher in male neonates 2 h after birth than at 0 h in both cesarean-delivered (9.48 +/- 1.25 vs. 3.37 +/- 0.60 ng/ml) and naturally delivered (19.28 +/- 2.94 vs. 5.13 +/- 1.93 ng/ml) ferrets, while female neonates showed no significant changes in T over these sampling times. T levels had returned to 0 h levels by 12 h in naturally delivered males. T was significantly lower in females than in males 0, 2, and 24 h after natural delivery, whereas T levels were equivalent in males and females immediately after cesarean delivery. Male kits kept on a heating pad for 2 h after natural delivery had lower plasma T levels than males that were left with their mothers over this same period. Brain aromatase activity in anterior hypothalamus-preoptic area, medial basal hypothalamus (MBH), temporal lobe, and cerebral cortex was equivalent in males and females at all postpartum ages, regardless of whether delivery occurred by cesarean section or naturally. However, in naturally delivered kits of both sexes significant elevations in aromatase activity occurred in MBH and temporal lobe 24 h postpartum. Finally, E2 concentrations in anterior hypothalamus-preoptic area and MBH were equivalent 0 and 2 h postpartum in males and females, regardless of whether they were delivered naturally or by cesarean section. The observed postnatal elevation in T may contribute to brain and behavioral sexual differentiation of male ferrets. It is unclear, however, whether such an effect of T depends on its neural aromatization to E2.


Assuntos
Animais Recém-Nascidos/fisiologia , Aromatase/metabolismo , Encéfalo/enzimologia , Carnívoros/fisiologia , Estradiol/análise , Furões/fisiologia , Hipotálamo/análise , Testosterona/sangue , Animais , Feminino , Masculino , Valores de Referência , Caracteres Sexuais
8.
Endocrinology ; 141(3): 1254-62, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10698203

RESUMO

GnRH neurons originate in the nasal compartment and migrate along vomeronasal fibers over the cribiform plate to the forebrain. Previously, we found gamma-aminobutyric acid (GABA) present in GnRH neurons during development. To clarify the influence of GABA across the entire GnRH migration route, we examined the effects of muscimol and bicuculline (GABA(A) agonist and antagonist) in vivo and in vitro, maintaining the integrity of the nasal-forebrain connection. For in vivo experiments, mice were administered muscimol, bicuculline, or vehicle on days 10-15 of pregnancy and were killed on embryonic day 15 (E15). For in vitro experiments, 250-microm parasagittal slices of whole heads of E13 mice were incubated with muscimol, bicuculline, or vehicle for 2 days. Muscimol inhibited GnRH cell migration and decreased extension of GnRH fibers. Bicuculline treatment led to a disorganized distribution of GnRH cells in the forebrain and a concomitant dissociation of GnRH cells from fibers of guidance. These results suggest that GABA's influence on GnRH development changes as the cells move out of the nasal compartment and extend processes toward the median eminence.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Neurônios/fisiologia , Receptores de GABA-A/efeitos dos fármacos , Animais , Bicuculina/farmacologia , Contagem de Células , Movimento Celular/efeitos dos fármacos , Feminino , Agonistas GABAérgicos/farmacologia , Antagonistas GABAérgicos/farmacologia , Agonistas de Receptores de GABA-A , Antagonistas de Receptores de GABA-A , Imuno-Histoquímica , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Muscimol/farmacologia , Fibras Nervosas/metabolismo , Hipófise/citologia , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Gravidez , Prosencéfalo/citologia , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/metabolismo
9.
Endocrinology ; 116(5): 1869-77, 1985 May.
Artigo em Inglês | MEDLINE | ID: mdl-3987621

RESUMO

Ferrets of both sexes were killed 8 or 5 days before expected parturition as well as 7, 15, 30, or 51 days after birth, and the activities of aromatase (using 19-[3H]hydroxyandrostenedione as substrate) and of 5 alpha-reductase (using [3H] testosterone as substrate) were assayed in whole homogenates of preoptic area plus anterior hypothalamus (POA), mediobasal hypothalamus (MBH), temporal lobe (TL), and cerebral cortex. Aromatase and testosterone 5 alpha-reductase activities were also measured in these regions in adult gonadectomized male and female ferrets. Compared with adults of both sexes in which aromatase activity was low in all brain regions studied, fetal ferrets had high levels of aromatase activity in POA plus MBH and in TL. At these prenatal ages, aromatase activity in POA plus MBH was significantly higher in males than in females. Aromatase activity in POA, MBH, and TL remained high in both sexes on postnatal days 7, 15, and 30, before declining by postnatal day 51. Cortical aromatase activity was uniformly low across all perinatal ages. The existence of a sex difference in aromatase activity in fetal POA plus MBH cannot be explained by a concurrent sex difference in circulating testosterone. Administration of testosterone to pregnant female ferrets over days 30-41 of gestation caused 150- to 350-fold increases in maternal plasma concentrations of testosterone and 2- to 5-fold increases in fetal plasma testosterone. However, aromatase activity was not affected in the POA and MBH of fetuses or mothers, although activity was significantly increased in the TL of mothers given testosterone. Furthermore, castration of neonatal or adult breeding males decreased plasma androgen levels by factors of 8 and 480, respectively, but resulted in only modest reductions in POA, MBH, and TL aromatase activity (a significant reduction occurred only in the adult male TL). Relatively high levels of testosterone 5 alpha-reductase activity were found in all brain regions across all perinatal ages, as well as in gonadectomized adult ferrets; there was no sex differences at any postnatal age studied. Prenatally, males had higher levels of 5 alpha-reductase activity than females only on day -8 in the POA plus MBH. The results show that estrogen and 5 alpha-reduced androgens can be synthesized in the brains of ferrets of both sexes during the perinatal period of sexual differentiation. A functional role for this neural metabolism of androgen remains to be demonstrated in this carnivorous species.


Assuntos
3-Oxo-5-alfa-Esteroide 4-Desidrogenase/metabolismo , Aromatase/metabolismo , Encéfalo/metabolismo , Feto/metabolismo , Oxirredutases/metabolismo , Testosterona/metabolismo , Fatores Etários , Análise de Variância , Animais , Feminino , Furões , Masculino , Gravidez , Fatores Sexuais
10.
Endocrinology ; 137(12): 5415-20, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8940365

RESUMO

Neurons containing the decapeptide GnRH originate in the olfactory placodes and migrate into the central nervous system during fetal development. The neurotransmitter gamma-aminobutyric acid (GABA) has been proposed as a trophic factor and may also influence neuronal migration. Immunocytochemical analyses were conducted in fetal rats, mice, and humans to identify potential developmental relationships between cells containing GABA, and GnRH neurons. Cells containing GABA were found along the nasal portion of the GnRH migration pathway in rats, mice, and humans during development. A peak number of cells containing immunoreactive GABA was observed in the nasal compartment of rats at embryonic day 15. At this time (E15), a majority of GnRH neurons were clustered in the region of the cribriform plate. By postnatal day 1, all GnRH neurons had migrated into the CNS and GABA cells were virtually absent from the nasal compartment. Double-label and confocal analyses of GABA and GnRH in mice and rats demonstrated that some olfactory GABAergic neurons coexpress GnRH. This implies that neurons that transiently express GABA originate in olfactory placodes and migrate into the forebrain. Based on the transient dual-label and adjacent relationships between GABA and GnRH containing cells in the nasal compartment, and other data showing migrational and trophic roles for GABA in development, we suggest that GABA may directly influence GnRH neuronal migration and development.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/fisiologia , Condutos Olfatórios/embriologia , Condutos Olfatórios/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Animais Recém-Nascidos/metabolismo , Movimento Celular/fisiologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário e Fetal , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Condutos Olfatórios/citologia , Ratos , Ratos Sprague-Dawley
11.
J Comp Neurol ; 332(3): 282-92, 1993 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-8331216

RESUMO

Sulfoglucuronylglycolipids (SGGLs) and glycoproteins, reacting with monoclonal antibody HNK-1, are developmentally and spatially regulated in the mammalian cortex and cerebellum. It has been proposed that the HNK-1 carbohydrate epitope is involved in intercellular adhesion and cell-cell interactions. Biochemical analysis and immunocytochemical localization of SGGLs and other neolacto series glycolipids were studied in the leaner mutant mouse cerebellum, where a slow and progressive rostral to caudal degeneration occurs with a gradual loss of both granule cells and Purkinje cells. Biochemical analyses showed that SGGLs and other neolacto series of glycolipids were significantly decreased in the adult leaner cerebellum; however, HNK-1-reactive glycoproteins were not affected. By an immunocytochemical method which selectively localizes the lipid antigens, it is shown that SGGLs are primarily associated with Purkinje cell bodies and their dendrites in the molecular layer and in cerebellar nuclei where Purkinje cell axons terminate. At postnatal day 30 (P30), SGGL immunoreactivity (SGGL-ir) in the leaner cerebellum was reduced moderately compared to normal littermates, which correlated with the minimal degree of Purkinje cell degeneration at this age in leaner and with the biochemical data. At P67 and P90, the SGGL-ir was significantly more reduced in the leaner as Purkinje cell degeneration proceeded. There was a direct correlation between loss of Purkinje cells and SGGL-ir in the cerebellar molecular layer. In both normal and young leaner cerebella, the SGGL-ir in different lobules was not uniform; there were distinct rostrocaudal and mediolateral differences. SGGL-ir was markedly more intense in rostral than in caudal lobules in the vermis, the dividing line being the region immediately caudal to the primary fissure and rostral to the declival sulcus. In the lateral cerebellum, the SGGL-ir was less intense than in the vermis and the rostrocaudal difference was not as pronounced. There was also nonuniformity in the intensity of staining in different folia. The rostrocaudal as well as mediolateral differences in the intensity of SGGL-ir were confirmed independently by biochemical analysis. The differential phenotypic expression of SGGLs and the selective susceptibility to Purkinje cell death in leaner mutant are discussed in relation to the known embryologic and ontogenetic compartmentation of cerebellum.


Assuntos
Anticorpos Monoclonais/imunologia , Cerebelo/metabolismo , Glicolipídeos/biossíntese , Mutação , Animais , Western Blotting , Moléculas de Adesão Celular Neuronais/imunologia , Moléculas de Adesão Celular Neuronais/metabolismo , Cerebelo/citologia , Cerebelo/crescimento & desenvolvimento , Gangliosídeos/metabolismo , Glicolipídeos/imunologia , Imuno-Histoquímica , Laminina/imunologia , Laminina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Células de Purkinje/metabolismo
12.
J Comp Neurol ; 376(1): 97-111, 1996 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-8946286

RESUMO

Gonadotropin releasing-hormone (GnRH) regulates the hypothalamo-pituitary-gonadal axis in vertebrates. The regulation of GnRH is intimately related to information from the olfactory system. Additionally, GnRH neurons are thought to be derived from progenitor cells in medial olfactory placodes. The present experiments were conducted to characterize the earliest development of GnRH neurons in lamprey and to determine their relationship to cells and fibers derived from the olfactory system. Eggs from fertile adult sea lamprey were fertilized in the laboratory, and larvae were maintained for up to 100 days. GnRH neurons were visualized within the lamprey preoptic area and hypothalamus as soon as GnRH was detectable (22 days after fertilization). The number of neurons increased with age through day 100. GnRH neurons were never seen within the olfactory system. The cells and fibers of the olfactory system were identified using the lectin, Grifonia Simplicifolia-1 (GS-1). Overlap between the olfactory and GnRH systems were at the level of fiber projections. GS-1 reactive cells of apparent placodal origin did not enter the region of the preoptic area or hypothalamus that contained GnRH neurons. Recently divided cells were labeled with the thymidine analog, bromodeoxyuridine (BrdU). The positions of BrdU-labeled cells after different survival times suggest a predominant medial-lateral radial neuron migration with a small number in positions suggestive of migration between the olfactory epithelium and the telencephalic lobes. Regardless of survival time, these cells were always found close to their entry point into the brain, suggesting minimal rostral-caudal migration. Based on these results, we hypothesize that GnRH neurons in developing lamprey originate within proliferative zones of the diencephalon and not in the olfactory system. Based on the overlap of olfactory- and GnRH-containing fibers from prolarval stages to metamorphosis, olfactory stimuli may play a major role in the regulation of GnRH secretion in lamprey.


Assuntos
Envelhecimento/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Lampreias/fisiologia , Neurônios/fisiologia , Condutos Olfatórios/fisiologia , Animais , Divisão Celular , Glicoconjugados/metabolismo , Histocitoquímica , Lampreias/crescimento & desenvolvimento , Larva , Lectinas , Neurônios/citologia , Neurônios/metabolismo , Condutos Olfatórios/citologia
13.
J Comp Neurol ; 405(1): 88-98, 1999 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10022198

RESUMO

The ventromedial nucleus of the hypothalamus (VMH) is a key nucleus for regulating homeostatic, neuroendocrine, and behavioral functions. We conducted immunocytochemical analyses by using antisera directed against gamma-aminobutyric acid (GABA), its synthetic enzyme glutamic acid decarboxylase (GAD67), GABA-A receptor subunits (alpha2, beta3, epsilon), estrogen receptor-alpha, and Neuropeptide Y (NPY) in the region of the VMH in embryonic mice to identify potential patterning elements for VMH formation. Cells and fibers containing GABA and GAD67 encircled the primordial VMH as early as embryonic day 13 (E13) when the cytoarchitecture of the VMH was not recognizable by Nissl stain. At E16-17 the cytoarchitecture of the VMH became recognizable by Nissl stain as GABAergic fibers invaded the nucleus, continued postnatally, and by adulthood the density of GABAergic fibers was greater inside than outside the VMH. GABA-A receptor subunit expression (beta3 by E13 and alpha2 by E15) within the primordial VMH suggested potential sensitivity to the surrounding GABA signal. Brain slices were used to test whether fibers from distal or proximal sites influenced VMH development. Coronal Vibratome slices were prepared and maintained in vitro for 0-3 days. Nissl stain analyses showed a uniform distribution of cells in the region of the VMH on the day of plating (E15). After 3 days in vitro, cellular aggregation suggesting VMH formation was seen. Nuclear formation in vitro suggests that key factors resided locally within the coronal plane of the slices. It is suggested that either GABA intrinsic to the region nearby the VMH directly influences the development and organization of the VMH, or along with other markers provides an early indicator of pattern determination that precedes the cellular organization of the VMH.


Assuntos
Hipotálamo Médio/química , Ácido gama-Aminobutírico/análise , Animais , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário e Fetal/fisiologia , Receptor alfa de Estrogênio , Glutamato Descarboxilase/análise , Hipotálamo Médio/embriologia , Imuno-Histoquímica , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeo Y/análise , Receptores de Estrogênio/análise , Receptores de GABA-A/análise
14.
J Comp Neurol ; 389(2): 277-88, 1997 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-9416922

RESUMO

A sexually dimorphic male nucleus (MN) is seen in Nissl-stained sections from the dorsal preoptic area/anterior hypothalamus (dPOA/AH) of male, but not female, ferrets. We used immunohistochemical methods to determine whether particular neuropeptides are found in cells of the MN. A sexually dimorphic cluster of galanin-immunoreactive (IR) cells was found in the dPOA/AH of ferrets killed either on embryonic day (E) 38 or in adulthood. Significantly more galanin-IR cells were distributed in the MN and in other subregions of the dPOA/AH of intact breeding males than estrous females. The density of galanin-IR cells in the dPOA/AH was significantly reduced in adult males by castration and restored to the level of intact breeding males by daily injections of testosterone propionate (TP) for 5 weeks. The same TP treatment failed to augment the density of galanin-IR cells in the dPOA/AH of adult, ovariectomized females. Computer-assisted image analysis and grid-crossing analysis showed that the area and the number of galanin-IR fibers in the dPOA/AH were significantly greater in adult females than in males, regardless of subjects' concurrent steroidal condition. A cluster of galanin-IR cells was present in the dPOA/AH of males, but not females, killed on either E34 or E38. Administration of TP between E28 and E37 significantly increased the density of galanin-IR cells in the dPOA/AH of females killed on E38, up to the level seen in control males. The results suggest that the capacity of cells located in the dPOA/AH to express galanin after adult steroid exposure is sexually differentiated by the fetal action of testosterone, or its metabolite, estradiol, in males.


Assuntos
Furões/fisiologia , Galanina/biossíntese , Hipotálamo Anterior/fisiologia , Orquiectomia , Ovariectomia , Área Pré-Óptica/fisiologia , Animais , Embrião de Mamíferos , Feminino , Hipotálamo Anterior/citologia , Masculino , Área Pré-Óptica/citologia , Valores de Referência , Caracteres Sexuais
15.
J Comp Neurol ; 323(4): 577-85, 1992 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-1430324

RESUMO

A sexually dimorphic group of cells at the dorsal border of the preoptic/anterior hypothalamic area (POA/AH) of ferrets has been previously identified in Nissl-stained tissue. In this study, Golgi-stained tissue was examined in order 1) to determine whether sex differences exist in dendritic dimensions of neurons from this region, and 2) to assess the effects of adult androgen treatment on dendritic morphology in ferrets of both sexes. Brains from adult ferrets given daily injections of testosterone propionate (5 mg/kg body weight) or oil vehicle for 5 weeks after gonadectomy were impregnated by Golgi-Cox procedures. After sectioning at 120 microns, 78 multipolar neurons were selected from the sexually dimorphic POA/AH of 12 ferrets and reconstructed in three dimensions with the aid of a computer-assisted neuron tracing system. Large sex differences were observed in somal area and most aspects of dendritic morphology, including total length, number of branches, and total dendritic surface area. Androgen also appeared to accentuate dendritic arborization in both sexes, but this effect was weaker than the sex effect, more apparent in males than females, and restricted to fewer variables. The most statistically significant effects of adult androgen treatment in males were found for total dendritic surface area and percentage of fourth order dendrites, and in females, average dendritic thickness. These data show that strong sex differences exist in dendritic structure of neurons in the POA/AH, and suggest that alterations in levels of gonadal steroids in adulthood may promote synaptic remodeling in a region of the brain involved in the control of sexually dimorphic behaviors.


Assuntos
Núcleo Hipotalâmico Anterior/crescimento & desenvolvimento , Dendritos/efeitos dos fármacos , Furões/fisiologia , Neurônios/efeitos dos fármacos , Área Pré-Óptica/crescimento & desenvolvimento , Testosterona/farmacologia , Animais , Núcleo Hipotalâmico Anterior/ultraestrutura , Dendritos/ultraestrutura , Feminino , Furões/anatomia & histologia , Masculino , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/ultraestrutura , Orquiectomia , Ovariectomia , Área Pré-Óptica/ultraestrutura , Caracteres Sexuais
16.
J Comp Neurol ; 209(3): 287-300, 1982 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-6752217

RESUMO

The purpose of this study was to determine the distribution of luteinizing hormone-releasing hormone (LHRH) cells and pathways projecting to the median eminence and organum vasculosum of the lamina terminalis in the hypothalamus of the rat. Immunopositive LHRH was detected by the PAP method of immunocytochemistry on vibratome sections without embedding. Female rats were ovariectomized and treated with estradiol benzoate or implanted with estradiol capsules prior to sacrifice in order to minimize to variations in LH and ultimately to maximize hypothalamic LHRH content. Immunoreactive LHRH perikarya are diffusely aggregated across several nuclear groups: nucleus of the vertical limb of the diagonal band of Broca, medial septal nucleus, medians preoptic nucleus, rostral and medial preoptic areas, anterior hypothalamic area, and lateral and basal hypothalamic areas. The aggregate of LHRH cells when projected upon a horizontal plane resembles the form of a V bisected by the third ventricle. The apex of the V of the V is directed rostrally toward the midline nuclear groups whereas the ends of the V incline ventrally toward the base of the brain and the median eminence. The majority of LHRH cells are in the rostral portion of the V in preoptic and anterior hypothalamic areas. Few cells are present in the basal hypothalamus. The processes of LHRH cells form two diffuse fiber systems which are separated by the midline hypothalamic nuclei over most of their course and converge in the basal hypothalamus close to the median eminence. The more lateral fiber system forms part of the medial forebrain bundle, while the periventricular system is associated with the wall of the third ventricle. The dispersion of LHRH cells over many nuclear groups may allow for the integration of afferents from divergent regions of the neuraxis to mediate both tonic and phasic gonadotropin secretion in the rat.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Eminência Mediana/metabolismo , Animais , Mapeamento Encefálico , Feminino , Hipotálamo/anatomia & histologia , Técnicas Imunoenzimáticas , Sistema Límbico/anatomia & histologia , Eminência Mediana/anatomia & histologia , Fibras Nervosas/ultraestrutura , Vias Neurais/anatomia & histologia , Vias Neurais/metabolismo , Neurônios/ultraestrutura , Ratos , Ratos Endogâmicos
17.
J Comp Neurol ; 423(4): 579-89, 2000 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-10880989

RESUMO

The ventromedial nucleus of the hypothalamus (VMH) in mice first emerges as a histologically distinct cell cluster around embryonic day 17 (E17). The earliest known marker for cells destined to form the VMH is the orphan nuclear receptor, steroidogenic factor 1 (SF-1), which can be detected in the hypothalamic primordium by E11. Strikingly, the VMH is absent in newborn SF-1 knockout mice, suggesting that SF-1 is essential for the development of VMH neurons. We reported previously that the VMH can be identified before it emerges as a histologically distinct nucleus (i.e., at E13) by the exclusion of cells that are immunoreactive for both gamma-aminobutyric acid (GABA) and the synthetic enzyme, glutamic acid decarboxylase (GAD67). Subsequently, by E15, the developing VMH is demarcated further by cells that are immunoreactive for neuropeptide Y, estrogen receptor alpha (ERalpha), and galanin. It is noteworthy that the normal exclusion of GABA from the developing VMH is not seen in SF-1 knockout mice, and cells that are immunoreactive for neuropeptide Y, ERalpha, and galanin also are distributed aberrantly in this region. Thus, the absence of SF-1 profoundly affects the cellular architecture of the VMH from early stages in its formation. These data suggest that, directly or indirectly, SF-1 plays important roles in determining the distribution of cells in the mediobasal hypothalamus.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neurônios/metabolismo , Fatores de Transcrição/genética , Núcleo Hipotalâmico Ventromedial/embriologia , Animais , Animais Recém-Nascidos , Embrião de Mamíferos , Receptor alfa de Estrogênio , Fatores de Transcrição Fushi Tarazu , Proteínas de Homeodomínio , Camundongos , Camundongos Knockout , Fibras Nervosas/metabolismo , Fibras Nervosas/ultraestrutura , Neurônios/citologia , Fenótipo , Receptores Citoplasmáticos e Nucleares , Receptores de Estrogênio/metabolismo , Fator Esteroidogênico 1 , Núcleo Hipotalâmico Ventromedial/citologia , Núcleo Hipotalâmico Ventromedial/metabolismo
18.
Neuroscience ; 85(3): 759-71, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9639270

RESUMO

Sulfoglucuronyl carbohydrate is the terminal moiety of neolacto-oligosaccharides, expressed on several glycoproteins of the immunoglobulin superfamily involved in cell-cell recognition and on two glycolipids. Sulfoglucuronyl carbohydrate is temporally and spatially regulated in the developing nervous system. It appears to be involved in neural cell recognition and in cell adhesion processes through its interaction with specific proteins on cell surfaces. Previously we have characterized a specific sulfoglucuronyl carbohydrate-binding protein in rat brain. Sulfoglucuronyl carbohydrate binding protein-1 is structurally similar to a 30,000 mol. wt adhesive and neurite outgrowth promoting protein amphoterin [Rauvala and Pihlaskari (1987) J. biol. Chem. 262, p. 16,625]. The pattern of expression of sulfoglucuronyl carbohydrate binding protein-1 in developing rat nervous system was studied to understand the significance of its interaction with sulfoglucuronyl carbohydrate-bearing molecules. Biochemical analyses showed that the expression of sulfoglucuronyl carbohydrate binding protein-1 was developmentally regulated similarly to sulfoglucuronyl carbohydrate. Immunocytochemical localization of sulfoglucuronyl carbohydrate binding protein-1 and sulfoglucuronyl carbohydrate was performed by bright-field and fluorescent confocal laser scanning microscopy. In postnatal day 7 rat cerebellum, sulfoglucuronyl carbohydrate binding protein-1 was primarily associated with neurons of the external and internal granule cell layers. The sulfoglucuronyl carbohydrate binding protein-1 immunoreactivity was absent in Purkinje cell bodies and their dendrites in the molecular layer, as well as in Bergmann glial fibres and in white matter. In contrast, sulfoglucuronyl carbohydrate (reactive with HNK-1 antibody) was localized in processes surrounding granule neurons in the internal granule cell layer. Sulfoglucuronyl carbohydrate was also expressed in Purkinje neurons and their dendrites in the molecular layer and their axonal processes in the white matter. To a lesser extent Bergmann glial fibres were also positive for sulfoglucuronyl carbohydrate. In the cerebral cortex, at embryonic day 21, sulfoglucuronyl carbohydrate binding protein-1 was mainly observed in immature neurons of the cortical plate and subplate and dividing cells near the ventricular zone. Whereas, sulfoglucuronyl carbohydrate was strongly expressed in the fibres of the subplate and marginal zone. Sulfoglucuronyl carbohydrate was also found in the processes surrounding the sulfoglucuronyl carbohydrate binding protein-1-expressing neuronal cell bodies in the cortical plate and in ventricular zone. The specific localization of sulfoglucuronyl carbohydrate binding protein- in cerebellar granule neurons and neurons of the cerebral cortex was also confirmed by immunocytochemistry of the dissociated tissue cell cultures. The complementary localization of sulfoglucuronyl carbohydrate and sulfoglucuronyl carbohydrate binding protein-1, both in cerebral cortex and cerebellum, in apposing cellular structures indicate possible interaction between the two and signalling during the process of cell migration and arrest of migration.


Assuntos
Antígenos CD57/biossíntese , Carboidratos/biossíntese , Cerebelo/química , Córtex Cerebral/química , Glicoproteínas/análise , Animais , Anticorpos , Especificidade de Anticorpos , Química Encefálica/fisiologia , Antígenos CD57/análise , Carboidratos/análise , Cerebelo/citologia , Córtex Cerebral/citologia , Imunofluorescência , Globosídeos/análise , Globosídeos/biossíntese , Glucuronatos/análise , Glucuronatos/metabolismo , Glicoproteínas/imunologia , Glicoproteínas/metabolismo , Microscopia Confocal , Neurônios/química , Neurônios/metabolismo , Coelhos , Ratos , Ratos Sprague-Dawley
19.
J Histochem Cytochem ; 38(8): 1193-200, 1990 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1694876

RESUMO

Monoclonal antibody (MAb) HNK-1 recognizes a carbohydrate epitope present in certain glycolipids, glycoproteins, and proteoglycans. Five different fixation methods, together with biochemical analyses of the antigens, were evaluated to study immunocytochemical localization of this epitope in layers of adult rat cerebellum; 4% paraformaldehyde/0.5% cetylpyridinium chloride was found to be optimal for overall immunoreactivity, and the antigens were apparent in all cerebellar layers. To differentially localize HNK-1-reactive carbohydrate epitope on proteins vs lipids in cerebellar layers, we tested the effect of 0.2%, 2%, or 4% glutaraldehyde combined with 2% paraformaldehyde (GT/PF) on HNK-1 and other MAb-reactive protein and lipid antigens; 2% or 4% GT/PF significantly reduced or abolished immunoreactivity of MAb HNK-1 and 5F9 (reacting with microtubule-associated protein 2) with cerebellar proteins analyzed on Western blots, but did not decrease HNK-1 reactivity to lipid antigens on HPTLC blots. In cerebellar tissue sections, HNK-1 and 5F9 immunoreactivity was reduced after 2% or 4% GT/PF fixation. However, significant amounts of HNK-1 immunoreactivity remained in molecular layer and deep cerebellar nuclei. GT/PF fixation did not cause significant changes in immunoreactivity patterns of other carbohydrate lipid antigens, such as those that react with MAb A2B5, 7A, and WCC4. Therefore, carbohydrate epitope on lipids, as opposed to that on proteins, may be preferentially detectable by immunocytochemistry after fixation with 2% or 4% GT/PF. The selective localization of HNK-1-reactive carbohydrate in the molecular layer and deep cerebellar nuclei with 2% or 4% GT/PF fixation correlates well with the observed presence of HNK-1-reactive lipids in these areas but not in the granular layer and white matter, as determined by microdissection of the individual layers and biochemical analysis. The application of 2% or 4% GT/PF fixation as a general method for differentiating the same carbohydrate epitope on proteins vs lipids in immunocytochemistry for other tissues and other antibodies remains to be further evaluated.


Assuntos
Antígenos de Diferenciação/análise , Cerebelo/análise , Epitopos/análise , Fixadores , Glicolipídeos/imunologia , Glicoproteínas/imunologia , Animais , Anticorpos Monoclonais , Especificidade de Anticorpos , Western Blotting , Boroidretos , Antígenos CD57 , Feminino , Formaldeído , Glutaral , Imuno-Histoquímica , Ácido Periódico , Polímeros , Ratos , Ratos Endogâmicos
20.
Mol Cell Endocrinol ; 185(1-2): 173-84, 2001 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-11738807

RESUMO

Gonadotropin-releasing hormone (GnRH) regulates the hypothalamo-pituitary-gonadal (HPG) axis in all vertebrates studied. GnRH neurons that regulate the HPG axis are primarily derived from progenitor cells in the nasal compartment (NC) and migrate along olfactory system derived fibers across the cribriform plate to destinations in the forebrain. Across their long and uncommon migratory route many factors are likely important for their successful development. Several classes of molecules are being studied for their potential influences on migration, including those related to cell surface interactions (membrane receptors, adhesion molecules, extracellular matrix (ECM) molecules, etc.) and those related to communication across distances (neurotransmitters, peptides, chemoattractant or repellent molecules). Of the classes of molecules associated with cell surface interactions, glycoconjugates with terminal galactose, are temporally and spatially expressed on olfactory fibers that guide GnRH neurons and may play role(s) in migration. Of the molecules associated with communication across distances, the neurotransmitter gamma-aminobutyric acid (GABA) is associated with the GnRH migration pathway and influences the position and organization of GnRH neurons in vitro and in vivo. Furthermore, galactose-containing glycoconjugates and GABA are associated with GnRH neurons in species ranging from humans to lamprey. In mice and rats, GABA is found transiently within a subpopulation of GnRH neurons as they migrate through the NC. One of the key elements in considering regulators of GnRH neuron migration is the diversity of GnRH synthesizing cells. For example, only subpopulations of GnRH neurons also contain GABA, specific GABA receptors, or select glycoconjugates. Similarly, treatments that influence GnRH neuronal migration may only affect specific subsets and not the entire population. It is likely that we will not be able to characterize the migration of all GnRH neurons by a single factor. By combining molecular inquiries with genetic models, single cell analyses, and an in vitro migration model, we are beginning to decipher one of the most critical events in the establishment of the reproductive axis.


Assuntos
Desenvolvimento Embrionário e Fetal , Hormônio Liberador de Gonadotropina/fisiologia , Animais , Movimento Celular/fisiologia , Humanos , Neurônios/química , Neurônios/citologia , Condutos Olfatórios/citologia , Condutos Olfatórios/embriologia , Ácido gama-Aminobutírico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA