Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Physiol Rep ; 11(22): e15873, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37994278

RESUMO

Organotypic lung slices, sometimes known as precision-cut lung slices (PCLS), provide an environment in which numerous cell types and interactions can be maintained outside the body (ex vivo). PCLS were maintained ex vivo for up to a week and demonstrated health via the presence of neurons, maintenance of tissue morphology, synthesis of mucopolysaccharides, and minimal cell death. Multiple phenotypes of neuronal fibers were present in lung slices with varied size, caliber, and neurotransmitter immunoreactivity. Of the neuropeptides present in fibers, calcitonin gene-related peptide (CGRP) was the most prevalent. Exposing PCLS to recombinant CGRP resulted in the proliferation and dispersion of CD19+ B cells in slices taken selectively from females. The number of granules containing immunoreactive (ir) surfactant protein C (SPC), which are representative of alveolar type 2 cells, increased in slices from females within 24 h of exposure to CGRP. Additionally, ir-SPC granule size increased in slices from males and females across 48 h of exposure to CGRP. Exposure of PCLS to exogenous CGRP did not alter the number of solitary pulmonary neuroendocrine cells (PNEC) but did result in neuroendocrine bodies that had significantly more cells. Neuronal fiber numbers were unchanged based on ir-peripherin; however, ir-CGRP became non-detectable in fibers while unchanged in PNECs. The effects of exogenous CGRP provide insight into innate immune and neuroendocrine responses in the lungs that may be partially regulated by neural fibers. The sex-dependent nature of these changes may point to the basis for sex-selective outcomes among respiratory diseases.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina , Neuropeptídeos , Masculino , Feminino , Humanos , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Neuropeptídeos/farmacologia , Neuropeptídeos/metabolismo , Pulmão/metabolismo , Neurônios/metabolismo , Tórax
2.
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
3.
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
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.
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
6.
J Neuroendocrinol ; 24(1): 225-35, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21668533

RESUMO

Steroidogenic factor 1 (SF-1; officially designated NR5a1) is a member of a nuclear receptor superfamily with important roles in the development of endocrine systems. Studies with global and tissue-specific (i.e. central nervous system) knockout mice have revealed several roles of SF-1 in brain. These include morphological effects on the development of the ventromedial nucleus of the hypothalamus and functional effects on body weight regulation through modulation of physical activity, anxiety-like behaviours and female sexual behaviours. Although such defects are almost certainly a result of the absence of SF-1 acting as a transcription factor in the hypothalamus, global SF-1 knockout mice also represent a model for studying the sex differences in the brain that develop in the absence of exposure to foetal sex steroid hormones as a result of the absence of gonads. In the present review, current knowledge of the roles of SF-1 protein in the central nervous system is discussed.


Assuntos
Sistema Nervoso Central/metabolismo , Fator Esteroidogênico 1/metabolismo , Animais , Feminino , Masculino , Caracteres Sexuais , Comportamento Sexual Animal/fisiologia
7.
Front Neuroendocrinol ; 31(3): 341-58, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20457175

RESUMO

Mice and rats are important mammalian models in biomedical research. In contrast to other biomedical fields, work on sexual differentiation of brain and behavior has traditionally utilized comparative animal models. As mice are gaining in popularity, it is essential to acknowledge the differences between these two rodents. Here we review neural and behavioral sexual dimorphisms in rats and mice, which highlight species differences and experimental gaps in the literature, that are needed for direct species comparisons. Moving forward, investigators must answer fundamental questions about their chosen organism, and attend to both species and strain differences as they select the optimal animal models for their research questions.


Assuntos
Encéfalo/fisiologia , Diferenciação Sexual/fisiologia , Comportamento Sexual Animal/fisiologia , Animais , Comportamento Animal/fisiologia , Encéfalo/crescimento & desenvolvimento , Humanos , Camundongos , Modelos Animais , Ratos , Especificidade da Espécie
8.
J Neuroendocrinol ; 22(6): 518-26, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20236232

RESUMO

Developmental exposure to the agricultural fungicide vinclozolin can impair reproductive function in male rabbits and was previously found to decrease the number of immunoreactive-gonadotrophin-releasing hormone (GnRH) neurones in the region of the organum vasculosum of the lamina terminalis and rostral preoptic area by postnatal week (PNW) 6. In the present study, in an aim to further examine the disruption of GnRH neurones by foetal vinclozolin exposure, pregnant rabbits were dosed orally with vinclozolin, flutamide or carrot paste vehicle for the last 2 weeks of gestation. Offspring were euthanised at birth (males and females), PNW 6 (females), PNW 26 (adult males) or PNW 30 (adult females) of age. At birth and in adults, brains were sectioned and processed for immunoreactive GnRH. The numbers of immunoreactive GnRH neuronal perikarya were significantly decreased in vinclozolin-treated rabbits at birth and in adult littermates. By contrast, there was an increase in GnRH immunoreactivity in the terminals in the region of the median eminence. Analysis of PNW 6 female brains by radioimmunoassay revealed a two-fold increase in GnRH peptide content in the mediobasal hypothalamus in vinclozolin-treated rabbits. This finding was complemented by immunofluorescence analyses, which revealed a 2.8-fold increase in GnRH immunoreactivity in the median eminence of vinclozolin compared to vehicle-treated females at PNW 30. However, there was no difference between treatment groups in the measures of reproduction that were evaluated: ejaculation latency, conception rates or litter size. These results indicate that sub-acute, prenatal vinclozolin treatment is sufficient to create perdurable alterations in the GnRH neuronal network that forms an important input into the reproductive axis. Finally, the effect of vinclozolin on the GnRH neuronal network was not comparable to that of flutamide, suggesting that vinclozolin was not acting through anti-androgenic mechanisms.


Assuntos
Fungicidas Industriais/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/efeitos dos fármacos , Oxazóis/farmacologia , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Imuno-Histoquímica , Masculino , Microscopia de Fluorescência , Neurônios/metabolismo , Gravidez , Coelhos , Radioimunoensaio , Reprodução
9.
J Neuroendocrinol ; 21(4): 387-92, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19207813

RESUMO

Sex differences in the nervous system come in many forms. Although a majority of sexually dimorphic characteristics in the brain have been described in older animals, mechanisms that determine sexually differentiated brain characteristics often operate during critical perinatal periods. Both genetic and hormonal factors likely contribute to physiological mechanisms in development to generate the ontogeny of sexual dimorphisms in brain. Relevant mechanisms may include neurogenesis, cell migration, cell differentiation, cell death, axon guidance and synaptogenesis. On a molecular level, there are several ways to categorize factors that drive brain development. These range from the actions of transcription factors in cell nuclei that regulate the expression of genes that control cell development and differentiation, to effector molecules that directly contribute to signalling from one cell to another. In addition, several peptides or proteins in these and other categories might be referred to as 'biomarkers' of sexual differentiation with undetermined functions in development or adulthood. Although a majority of sex differences are revealed as a direct consequence of hormone actions, some may only be revealed after genetic or environmental disruption. Sex differences in cell positions in the developing hypothalamus, and steroid hormone influences on cell movements in vitro, suggest that cell migration may be one target for early molecular actions that impact brain development and sexual differentiation.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiologia , Movimento Celular/fisiologia , Estrogênios/metabolismo , Caracteres Sexuais , Esteroides/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Feminino , Humanos , Masculino , Neurônios/fisiologia , Óxido Nítrico/metabolismo , Diferenciação Sexual/fisiologia , Transdução de Sinais , Ácido gama-Aminobutírico/metabolismo
10.
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
11.
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
12.
J Neurobiol ; 49(4): 264-76, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11745664

RESUMO

The region that becomes the ventromedial nucleus of the hypothalamus (VMH) is surrounded by cells and fibers containing immunoreactive gamma-aminobutyric acid (GABA) by embryonic day 13 (E13), several days before the nucleus emerges in Nissl stains. As GABA plays many roles during neural development, we hypothesized that it influences VMH development, perhaps by providing boundary information for migrating neurons. To test this hypothesis we examined the VMH in embryonic mice in which the beta3 subunit of the GABA(A)-receptor, a receptor subunit that is normally highly expressed in this nucleus, was disrupted by gene targeting. In beta3 -/- embryos the VMH was significantly larger, and the distribution of cells containing immunoreactive estrogen receptor-alpha was expanded compared to controls. Using in vitro brain slices from wild-type C57BL/6J mice killed at E15 we found that treatment with the GABA(A) antagonist bicuculline increased the number of cells migrating per video field analyzed in the VMH. In addition, treatment with either bicuculline or the GABA(A) agonist muscimol altered the orientation of cell migration in particular regions of this nucleus. These data suggest that GABA is important for the organization of cells during VMH formation.


Assuntos
Agonistas de Receptores de GABA-A , Antagonistas de Receptores de GABA-A , Núcleo Hipotalâmico Ventromedial/embriologia , Ácido gama-Aminobutírico/fisiologia , Animais , Química Encefálica/genética , Receptor alfa de Estrogênio , Feminino , Corantes Fluorescentes , Agonistas GABAérgicos/farmacologia , Antagonistas GABAérgicos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Imuno-Histoquímica , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Vídeo , Gravidez , Receptores de Estrogênio/metabolismo , Receptores de GABA-A/genética , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos , Núcleo Hipotalâmico Ventromedial/fisiologia
13.
Horm Behav ; 40(2): 86-92, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11534967

RESUMO

The interstitial nuclei of the human anterior hypothalamus (INAH1-4) have been considered candidates for homology with the sexually dimorphic nucleus of the preoptic area of the rat. Volumetric sexual dimorphism has been described for three of these nuclei (INAH1-3), and INAH3 has been reported to be smaller in homosexual than heterosexual men. The current study measured the INAH in Nissl-stained coronal sections in autopsy material from 34 presumed heterosexual men (24 HIV- and 10 HIV+), 34 presumed heterosexual women (25 HIV- and 9 HIV+), and 14 HIV+ homosexual men. HIV status significantly influenced the volume of INAH1 (8% larger in HIV+ heterosexual men and women relative to HIV- individuals), but no other INAH. INAH3 contained significantly more neurons and occupied a greater volume in presumed heterosexual males than females. No sex difference in volume was detected for any other INAH. No sexual variation in neuronal size or density was observed in any INAH. Although there was a trend for INAH3 to occupy a smaller volume in homosexual men than in heterosexual men, there was no difference in the number of neurons within the nucleus based on sexual orientation.


Assuntos
Infecções por HIV/patologia , Hipotálamo/fisiologia , Comportamento Sexual/fisiologia , Adulto , Idoso , Encéfalo/anatomia & histologia , Encéfalo/patologia , Feminino , Soronegatividade para HIV/fisiologia , Soropositividade para HIV/patologia , Humanos , Hipotálamo/anatomia & histologia , Hipotálamo/patologia , Masculino , Pessoa de Meia-Idade , Tamanho do Órgão/fisiologia , Caracteres Sexuais
14.
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
15.
J Neurosci Res ; 62(2): 186-205, 2000 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11020212

RESUMO

Developmental expression of sulfoglucuronyl carbohydrate (SGC) and its binding protein, SBP-1 was studied in the rat cerebral cortex to understand their function. Between embryonic day (ED) 14-19, SBP-1 was strongly expressed in neurons of the ventricular zone and migrating neurons throughout the cortex. SBP-1 declined at birth and by postnatal day (PD) 3 only the latest arriving neurons in the most superficial segment of the cortical plate expressed SBP-1. Between ED 14-16, SGC was expressed in a thin row of glial cells near the ventricles and on their radial processes. Between ED 16-PD 3, SGC was not in neuronal cell soma, but was in neuronal plasma membranes and processes surrounding the neuronal perikarya. The expression of SGC declined similar to SBP-1 and both of them disappeared by PD 7. The expression of SBP-1 and SGC was chronologically coordinated with neuronal migration. SBP-1 was specifically expressed in immature neuronal nuclei and plasma membranes. SBP-1 and SGC were colocalized and were available for interaction with each other on neuronal cell membranes and processes. This was confirmed with isolated neurons in culture. As in vivo, the expression of SBP-1 in neurons declined with time in culture. The dissociated cortical neurons when plated on SBP-1 as a substratum produced extensive neuritic outgrowth. HNK-1, anti-SBP-1 antibodies and sulfoglucuronyl glycolipid, SGGL specifically and severely reduced neurite outgrowth. SBP-1-SGC interactions provide a potential mechanism for guidance and cell signaling, in the processes of neuronal migration and terminal differentiation.


Assuntos
Antígenos CD57/metabolismo , Metabolismo dos Carboidratos , Movimento Celular/fisiologia , Córtex Cerebral/metabolismo , Glicolipídeos/metabolismo , Glicoproteínas/metabolismo , Neurônios/metabolismo , Animais , Animais Recém-Nascidos , Anticorpos Monoclonais , Antígenos CD57/imunologia , Adesão Celular , Células Cultivadas , Córtex Cerebral/crescimento & desenvolvimento , Embrião de Mamíferos , Epitopos , Feminino , Neuritos/metabolismo , Gravidez , Ligação Proteica , Ratos , Ratos Sprague-Dawley
16.
J Biol Chem ; 275(50): 39662-70, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-10995769

RESUMO

Synthesis of the hypothalamic peptide, gonadotropin releasing hormone (GnRH), is paramount for reproductive function. GnRH neurons originate in the olfactory region and migrate into the forebrain during development. We recently implicated adhesion related kinase (Ark) in GnRH neuron development based on its differential expression in two GnRH producing cell lines, GT1-7 and Gn10. The Ark membrane receptor encodes an extracellular domain resembling cell adhesion molecules and an intracellular tyrosine kinase. Ark is expressed in Gn10 cells derived from migrating GnRH neurons but not GT1-7 cells of the post-migratory phenotype. Here, we show that Ark and GnRH transcripts are colocalized in the cribriform plate at midgestation, suggesting that Ark is expressed in migrating GnRH neurons in vivo. Furthermore, we have identified the GnRH gene as a downstream target of Ark signaling. Ark inhibits GnRH gene expression in GnRH neuronal cells via the coordinated binding of myocyte enhancer factor-2B and -2C (MEF-2B and -2C) and a putative homeoprotein within the proximal rat GnRH promoter. Given that MEF-2 proteins are widely expressed in the brain, these studies provide further evidence for MEF-2 action during neuronal development. Moreover, our studies elucidate a potential role for Ark in regulating GnRH gene expression during GnRH neuronal migration.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Fatores de Regulação Miogênica/fisiologia , Proteínas Oncogênicas , Receptores Proteína Tirosina Quinases/metabolismo , Fatores de Transcrição/fisiologia , Animais , Sequência de Bases , Northern Blotting , Western Blotting , Encéfalo/metabolismo , Linhagem Celular , Movimento Celular , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Eletroforese em Gel de Poliacrilamida , Genes Dominantes , Fatores de Transcrição MEF2 , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fatores de Regulação Miogênica/biossíntese , Fatores de Regulação Miogênica/genética , Neurônios/metabolismo , Fenótipo , Regiões Promotoras Genéticas , Prosencéfalo/metabolismo , Ligação Proteica , Proteínas Tirosina Quinases/química , Proteínas Proto-Oncogênicas , Ratos , Transdução de Sinais , Fatores de Tempo , Distribuição Tecidual , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Transfecção , Receptor Tirosina Quinase Axl
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.
Brain Res Dev Brain Res ; 120(2): 165-80, 2000 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-10775769

RESUMO

Sulfoglucuronyl carbohydrate (SGC) is expressed on several glycoproteins of the immunoglobulin superfamily of cell-adhesion molecules. Developmental expression of SGC and its binding protein, SBP-1, was studied in the rat cerebellum by immunocytochemistry to understand the function of SBP-1 and the significance of its interaction with SGC. During early postnatal development (postnatal day (PD) 3-10) SBP-1 was strongly expressed in the granule neurons of the external and internal granule cell layers (EGCL and IGCL). This expression declined by PD 15, and disappeared in the adult. Between PD 3 and 15, SGC was expressed in cellular processes surrounding the granule neurons in the IGCL, and it also declined and disappeared with development. SGC expression, however, continued in Purkinje cells and their dendrites in the molecular layer in adults. The expressions of SBP-1 and SGC were developmentally regulated and appeared to be chronologically co-ordinated with granule neuron migration from EGCL to IGCL. High magnification confocal microscopy showed that SBP-1 was primarily localized in nuclei and plasma membranes of granule neurons, whereas SGC in the IGCL was localized on neuronal plasma membranes, dendrites and glial processes, but not in cell soma. The relative localization of SBP and SGC was confirmed by cellular and subcellular markers in vivo and with dissociated cerebellar cells in culture. It is proposed that SBP-1 on plasma membranes of granule neurons interacts with SGC on the surrounding processes and membranes and this interaction could provide a potential mechanism for guidance and cell signaling, in the processes of granule neuron migration and differentiation.


Assuntos
Antígenos CD57/metabolismo , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Envelhecimento/metabolismo , Animais , Animais Recém-Nascidos , Adesão Celular/fisiologia , Comunicação Celular/fisiologia , Compartimento Celular/fisiologia , Membrana Celular/metabolismo , Células Cultivadas , Cerebelo/citologia , Imuno-Histoquímica , Microscopia Confocal , Proteínas Associadas aos Microtúbulos/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Neurônios/citologia , Ligação Proteica , Ratos , Ratos Sprague-Dawley
19.
Gen Comp Endocrinol ; 117(3): 381-94, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10764549

RESUMO

The present study investigated GnRH forms within the brain of a representative of the order Cypriniformes, the white sucker, Catostomus commersoni, using HPLC, RIA, and immunocytochemistry. Several immunoreactive (ir) GnRH forms were identified in the brain of the white sucker by chromatography and radioimmunoassay, including ir-salmon GnRH, ir-lamprey GnRH-I and -III, and ir-chicken GnRH-II. Results from immunocytochemical studies were consistent with multiple GnRH forms distributed in different patterns, particularly for fibers. Neuronal perikarya containing ir-salmon GnRH and ir-lamprey-like GnRH were found laterally within the preoptic area and rostral hypothalamus. Cells containing exclusively ir-salmon GnRH appeared slightly more rostrally, but in the same region. Fibers containing ir-salmon GnRH and ir-lamprey-like GnRH were seen throughout the caudal telencephalon and extended into the diencephalon, toward the pituitary. Fibers containing ir-chicken-II-like GnRH were also seen in the caudal telencephalon, but were concentrated more dorsally in the diencephalon. Within the pituitary, fibers containing ir-salmon GnRH and ir-lamprey-like GnRH entered the neurohypophysis, but differed in their destinations. Fibers containing ir-salmon GnRH remained within the neurohypophysis, while fibers containing ir-lamprey-like GnRH targeted adenohypophyseal tissue. These findings are consistent with the hypothesis that multiple GnRH forms with multiple functions exist within the brain and pituitary of teleosts and provide further evidence of a lamprey-like GnRH within an early evolved teleost species.


Assuntos
Química Encefálica , Cipriniformes/metabolismo , Hormônio Liberador de Gonadotropina/análise , Sequência de Aminoácidos , Animais , Cromatografia Líquida de Alta Pressão , Diencéfalo/química , Hipotálamo/química , Imuno-Histoquímica , Lampreias , Área Pré-Óptica/química , Radioimunoensaio , Salmão , Homologia de Sequência , Telencéfalo/química
20.
Brain Res ; 856(1-2): 142-51, 2000 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-10677621

RESUMO

The distribution of estrogen receptor protein-alpha (ER-alpha)-containing cells in the human hypothalamus and adjacent regions was studied using a monoclonal antibody (H222) raised against ER-alpha derived from MCF-7 human breast cancer cells. Reaction product was found in restricted populations of neurons and astrocyte-like cells. Neurons immunoreactive for ER-alpha were diffusely distributed within the basal forebrain and preoptic area, infundibular region, central hypothalamus, basal ganglia and amygdala. Immunoreactive astrocyte-like cells were noted within specific brain regions, including the lamina terminalis and subependymal peri-third-ventricular region. These data are consistent with the location of estrogen receptors in the basal forebrain of other species and the known effects of estrogens on the cellular functions of both neurons and supporting elements within the human hypothalamus and basal forebrain.


Assuntos
Astrócitos/citologia , Encéfalo/citologia , Hipotálamo/citologia , Neurônios/citologia , Prosencéfalo/citologia , Receptores de Estrogênio/análise , Tonsila do Cerebelo/citologia , Anticorpos Monoclonais , Neoplasias da Mama , Receptor alfa de Estrogênio , Feminino , Globo Pálido/citologia , Humanos , Imuno-Histoquímica , Área Pré-Óptica/citologia , Substância Inominada/citologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...