Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters








Publication year range
1.
Front Neuroendocrinol ; 35(1): 140-58, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24355523

ABSTRACT

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.


Subject(s)
Cardiovascular Diseases/metabolism , Depression/metabolism , Fetal Development/physiology , Sex Characteristics , Stress, Physiological , Animals , Cardiovascular Diseases/physiopathology , Depression/physiopathology , Female , Humans , Male , Risk
2.
Pflugers Arch ; 465(5): 585-94, 2013 May.
Article in English | MEDLINE | ID: mdl-23503726

ABSTRACT

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.


Subject(s)
Cardiovascular Diseases/physiopathology , Depressive Disorder, Major/physiopathology , Sex Characteristics , Brain/metabolism , Cardiovascular Diseases/epidemiology , Comorbidity , Depressive Disorder, Major/epidemiology , Female , Humans , Male
3.
Horm Metab Res ; 44(8): 577-86, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22700441

ABSTRACT

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.


Subject(s)
Epigenesis, Genetic , Growth and Development/physiology , Hormones/metabolism , Aging/genetics , Animals , Brain/growth & development , Brain/metabolism , Growth and Development/genetics , Humans , Reproduction/genetics
4.
Horm Metab Res ; 44(8): 619-24, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22488519

ABSTRACT

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.


Subject(s)
Blood Vessels/growth & development , Blood Vessels/metabolism , Paraventricular Hypothalamic Nucleus/anatomy & histology , Paraventricular Hypothalamic Nucleus/blood supply , Signal Transduction , gamma-Aminobutyric Acid/metabolism , Animals , Blood Vessels/anatomy & histology , Mice , Mice, Inbred C57BL , Paraventricular Hypothalamic Nucleus/metabolism , Receptors, GABA-B/metabolism
5.
J Neuroendocrinol ; 24(1): 225-35, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21668533

ABSTRACT

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.


Subject(s)
Central Nervous System/metabolism , Steroidogenic Factor 1/metabolism , Animals , Female , Male , Sex Characteristics , Sexual Behavior, Animal/physiology
6.
J Neuroendocrinol ; 22(6): 518-26, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20236232

ABSTRACT

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.


Subject(s)
Fungicides, Industrial/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Neurons/drug effects , Oxazoles/pharmacology , Prenatal Exposure Delayed Effects , Animals , Female , Immunohistochemistry , Male , Microscopy, Fluorescence , Neurons/metabolism , Pregnancy , Rabbits , Radioimmunoassay , Reproduction
7.
Neuroscience ; 151(4): 1119-31, 2008 Feb 19.
Article in English | MEDLINE | ID: mdl-18248902

ABSTRACT

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.


Subject(s)
Cell Movement/physiology , Neurons/physiology , Receptors, GABA-B/physiology , Ventromedial Hypothalamic Nucleus/cytology , Animals , Animals, Newborn , Baclofen/analogs & derivatives , Baclofen/pharmacology , Cell Movement/drug effects , Dose-Response Relationship, Drug , Embryo, Mammalian , Estrogen Receptor alpha/metabolism , GABA Antagonists/pharmacology , In Vitro Techniques , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Protein Subunits/metabolism , Receptors, GABA-A/metabolism , Receptors, GABA-B/deficiency , Ventromedial Hypothalamic Nucleus/embryology , Ventromedial Hypothalamic Nucleus/growth & development
8.
J Neurobiol ; 49(4): 264-76, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11745664

ABSTRACT

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.


Subject(s)
GABA-A Receptor Agonists , GABA-A Receptor Antagonists , Ventromedial Hypothalamic Nucleus/embryology , gamma-Aminobutyric Acid/physiology , Animals , Brain Chemistry/genetics , Estrogen Receptor alpha , Female , Fluorescent Dyes , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Inbred C57BL , Microscopy, Video , Pregnancy , Receptors, Estrogen/metabolism , Receptors, GABA-A/genetics , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/physiology
9.
Mol Cell Endocrinol ; 185(1-2): 173-84, 2001 Dec 20.
Article in English | MEDLINE | ID: mdl-11738807

ABSTRACT

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.


Subject(s)
Embryonic and Fetal Development , Gonadotropin-Releasing Hormone/physiology , Animals , Cell Movement/physiology , Humans , Neurons/chemistry , Neurons/cytology , Olfactory Pathways/cytology , Olfactory Pathways/embryology , gamma-Aminobutyric Acid/physiology
10.
J Neurosci ; 21(3): 911-9, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11157077

ABSTRACT

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.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Movement/physiology , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Prosencephalon/metabolism , Tumor Suppressor Proteins , Animals , Animals, Newborn , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/pharmacology , Cell Count , Cell Movement/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , DCC Receptor , Homozygote , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neurons/cytology , Neurons/drug effects , Olfactory Pathways/cytology , Olfactory Pathways/embryology , Olfactory Pathways/metabolism , Prosencephalon/cytology , Prosencephalon/embryology , RNA, Messenger/biosynthesis , Rats , Receptors, Cell Surface , Vomeronasal Organ/cytology , Vomeronasal Organ/embryology , Vomeronasal Organ/innervation , Vomeronasal Organ/metabolism
11.
J Neurosci Res ; 62(2): 186-205, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11020212

ABSTRACT

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.


Subject(s)
CD57 Antigens/metabolism , Carbohydrate Metabolism , Cell Movement/physiology , Cerebral Cortex/metabolism , Glycolipids/metabolism , Glycoproteins/metabolism , Neurons/metabolism , Animals , Animals, Newborn , Antibodies, Monoclonal , CD57 Antigens/immunology , Cell Adhesion , Cells, Cultured , Cerebral Cortex/growth & development , Embryo, Mammalian , Epitopes , Female , Neurites/metabolism , Pregnancy , Protein Binding , Rats , Rats, Sprague-Dawley
12.
J Biol Chem ; 275(50): 39662-70, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-10995769

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/physiology , Gonadotropin-Releasing Hormone/metabolism , Myogenic Regulatory Factors/physiology , Oncogene Proteins , Receptor Protein-Tyrosine Kinases/metabolism , Transcription Factors/physiology , Animals , Base Sequence , Blotting, Northern , Blotting, Western , Brain/metabolism , Cell Line , Cell Movement , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Electrophoresis, Polyacrylamide Gel , Genes, Dominant , MEF2 Transcription Factors , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/genetics , Neurons/metabolism , Phenotype , Promoter Regions, Genetic , Prosencephalon/metabolism , Protein Binding , Protein-Tyrosine Kinases/chemistry , Proto-Oncogene Proteins , Rats , Signal Transduction , Time Factors , Tissue Distribution , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection , Axl Receptor Tyrosine Kinase
13.
J Comp Neurol ; 423(4): 579-89, 2000 Aug 07.
Article in English | MEDLINE | ID: mdl-10880989

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental/physiology , Neurons/metabolism , Transcription Factors/genetics , Ventromedial Hypothalamic Nucleus/embryology , Animals , Animals, Newborn , Embryo, Mammalian , Estrogen Receptor alpha , Fushi Tarazu Transcription Factors , Homeodomain Proteins , Mice , Mice, Knockout , Nerve Fibers/metabolism , Nerve Fibers/ultrastructure , Neurons/cytology , Phenotype , Receptors, Cytoplasmic and Nuclear , Receptors, Estrogen/metabolism , Steroidogenic Factor 1 , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/metabolism
14.
Gen Comp Endocrinol ; 117(3): 381-94, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10764549

ABSTRACT

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.


Subject(s)
Brain Chemistry , Cypriniformes/metabolism , Gonadotropin-Releasing Hormone/analysis , Amino Acid Sequence , Animals , Chromatography, High Pressure Liquid , Diencephalon/chemistry , Hypothalamus/chemistry , Immunohistochemistry , Lampreys , Preoptic Area/chemistry , Radioimmunoassay , Salmon , Sequence Homology , Telencephalon/chemistry
15.
Endocrinology ; 141(3): 1254-62, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10698203

ABSTRACT

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.


Subject(s)
Gonadotropin-Releasing Hormone/physiology , Neurons/physiology , Receptors, GABA-A/drug effects , Animals , Bicuculline/pharmacology , Cell Count , Cell Movement/drug effects , Female , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , GABA-A Receptor Agonists , GABA-A Receptor Antagonists , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Muscimol/pharmacology , Nerve Fibers/metabolism , Pituitary Gland/cytology , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Pregnancy , Prosencephalon/cytology , Prosencephalon/drug effects , Prosencephalon/metabolism
16.
Brain Res ; 856(1-2): 142-51, 2000 Feb 21.
Article in English | MEDLINE | ID: mdl-10677621

ABSTRACT

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.


Subject(s)
Astrocytes/cytology , Brain/cytology , Hypothalamus/cytology , Neurons/cytology , Prosencephalon/cytology , Receptors, Estrogen/analysis , Amygdala/cytology , Antibodies, Monoclonal , Breast Neoplasms , Estrogen Receptor alpha , Female , Globus Pallidus/cytology , Humans , Immunohistochemistry , Preoptic Area/cytology , Substantia Innominata/cytology , Tumor Cells, Cultured
17.
J Neurobiol ; 41(2): 252-66, 1999 Nov 05.
Article in English | MEDLINE | ID: mdl-10512982

ABSTRACT

The preoptic area/anterior hypothalamus (POA/AH) sits as a boundary region rostral to the classical diencephalic hypothalamus and ventral to the telencephalic septal region. Numerous studies have pointed to the region's importance for sex-dependent functions. Previous studies suggested that migratory guidance cues within this region might be particularly unique in their diversity. To better understand the early development and differentiation of the POA/AH, cytoarchitectural, birthdate, immunocytochemical, and cell migration studies were conducted in vivo and in vitro using embryonic C57BL/6J mice. A medial preoptic nucleus became discernible using Nissl stain in males and females between embryonic days (E) E15 and E17. Cells containing immunoreactive estrogen receptor-alpha were detected in the POA/AH by E13, and increased in number with age in both sexes. From E15 to E17, examination of the radial glial fiber pattern by immunocytochemistry confirmed the presence of dual orientations for migratory guidance ventral to the anterior commissure (medial-lateral and dorsal-ventral) and uniform orientation more caudally (medial-lateral). Video microscopy studies followed the migration of DiI-labeled cells in coronal 250-microm brain slices from E15 mice maintained in serum-free media for 1-3 days. Analyses showed significant migration along a dorsal-ventral orientation in addition to medial-lateral. The video analyses showed significantly more medial-lateral migration in males than females in the caudal POA/AH. In vivo, changes in the distribution of cells labeled by the mitotic indicator bromodeoxyuridine (BrdU) suggested their progressive migration through the POA/AH. BrdU analyses also indicated significant movement from dorsal to ventral regions ventral to the anterior commissure. The significant dorsal-ventral migration of cells in the POA/AH provides additional support for the notion that the region integrates developmental information from both telencephalic and diencephalic compartments. The sex difference in the orientation of migration of cells in the caudal POA/AH suggests one locus for the influence of gonadal steroids in the embryonic mouse forebrain.


Subject(s)
Cell Movement/physiology , Neurons/cytology , Preoptic Area/cytology , Preoptic Area/embryology , Sex Characteristics , Animals , Female , Male , Mice , Mice, Inbred C57BL , Microscopy, Video , Neuroglia/physiology , Neurons/physiology , Pregnancy
18.
Brain Res Dev Brain Res ; 115(2): 171-82, 1999 Jun 02.
Article in English | MEDLINE | ID: mdl-10407134

ABSTRACT

While sex differences in neural morphology in the preoptic area/anterior hypothalamus (POA/AH) have been demonstrated in many species, their existence in mice have been controversial. Given the increased use of transgenic and gene-disrupted mice, we characterized sex differences using Nissl stains, and the immunocytochemical location of estrogen receptor-alpha (ER-alpha) and galanin in the POA/AH of two widely used strains, C57BL/6 and 129SvEv, and a mixed strain (C57BL/6x129Sv); the wild-type littermates of steroidogenic factor-1 (SF-1) gene-disrupted mice. Cell grouping was not a reliable marker of sex. In adults, cells located beneath the anterior commissure (AC) were reliably larger in females than males in 129SvEv, but not in the other strains. Caudally, cells in a group medial to the medial extension of the bed nucleus of the stria terminalis (BST) were significantly larger in males than females in C57BL/6J and SF-1 gene-disrupted wild-types. Cell groups discernible by embryonic day (E) 18 were not sexually dimorphic for cell size in C57BL/6J mice at E18 or postnatal day (P) 4. The pattern of distribution of cells containing ER-alpha was similar among the strains, reduced in the group medial to the BST; a pattern established by P0. Galanin-containing cells and fibers were seen from E15 to adulthood ventral to the AC. Caudally, a smaller group ventromedial to the BST was found only in 129SvEv adults. Sex differences in neural morphology which develop within the POA/AH depend upon multiple factors, particularly including genetic background.


Subject(s)
Hypothalamus, Anterior/physiology , Preoptic Area/physiology , Sex Characteristics , Animals , Embryonic and Fetal Development/physiology , Estrogen Receptor alpha , Female , Galanin/analysis , Hypothalamus, Anterior/embryology , Hypothalamus, Anterior/growth & development , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Preoptic Area/embryology , Preoptic Area/growth & development , Receptors, Estrogen/analysis , Species Specificity , Steroidogenic Factor 1
19.
Brain Res ; 826(2): 220-9, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10224299

ABSTRACT

Studies in fetal brain tissue of rodents, nonhuman primates and birds have demonstrated that cells containing gonadotropin-releasing hormone (GnRH) migrate from the olfactory placode across the nasal septum into the forebrain. The purpose of this study was to examine GnRH neurons in components of the adult and fetal human olfactory system. In the adult human brain (n=4), immunoreactive GnRH was evident within diffusely scattered cell bodies and processes in the olfactory bulb, olfactory nerve, olfactory cortex, and nervus terminalis located on the anterior surface of the gyrus rectus. GnRH-immunoreactive structures showed a similar distribution in 20-week human fetal brains (n=2), indicating that the migration of GnRH neurons is complete at this time. In 10-11-week fetal brains (n=2), more cells were noted in the nasal cavity than in the brain. Our data are consistent with observations made in other species, confirming olfactory derivation and migration of GnRH neurons into the brain from the olfactory placode.


Subject(s)
Gonadotropin-Releasing Hormone/analysis , Olfactory Bulb/chemistry , Olfactory Bulb/embryology , Olfactory Nerve/chemistry , Olfactory Nerve/embryology , Adult , Age Factors , Antibodies , Brain Chemistry/physiology , Cell Movement/physiology , Fetus/chemistry , Gonadotropin-Releasing Hormone/immunology , Humans , Immunoenzyme Techniques , Neurons/chemistry , Neurons/physiology , Olfactory Bulb/cytology , Olfactory Nerve/cytology
20.
J Comp Neurol ; 405(1): 88-98, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10022198

ABSTRACT

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.


Subject(s)
Hypothalamus, Middle/chemistry , gamma-Aminobutyric Acid/analysis , Animals , Embryo, Mammalian/metabolism , Embryonic and Fetal Development/physiology , Estrogen Receptor alpha , Glutamate Decarboxylase/analysis , Hypothalamus, Middle/embryology , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Inbred C57BL , Neuropeptide Y/analysis , Receptors, Estrogen/analysis , Receptors, GABA-A/analysis
SELECTION OF CITATIONS
SEARCH DETAIL