Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Am J Physiol Heart Circ Physiol ; 309(2): H335-44, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25980022

ABSTRACT

The role of serotonin in the hemodynamic response to blood loss remains controversial. Caudal raphe serotonin neurons are activated during hypotensive hemorrhage, and their destruction attenuates sympathetic increases following blood loss in unanesthetized rats. Caudal raphe neurons provide serotonin-positive projections to the nucleus tractus solitarii (NTS), and disruption of serotonin-positive nerve terminals in the NTS attenuates sympathetic recovery following hemorrhage. Administration of 5-HT1A-receptor agonists following hemorrhage augments sympathetic-mediated increases in venous tone and tissue hypoxia. These findings led us to hypothesize that severe blood loss promotes activation of 5-HT1A receptors in the NTS, which facilitates sympathetic recovery and peripheral tissue perfusion. Here, we developed an adeno-associated viral vector encoding an efficacious small hairpin RNA sequence targeting the rat 5-HT1A receptor. Unanesthetized rats subjected to NTS injection of the anti-rat 5-HT1A small hairpin RNA-encoding vector 4 wk prior showed normal blood pressure recovery, but an attenuated recovery of renal sympathetic nerve activity (-6.4 ± 12.9 vs. 42.6 ± 15.6% baseline, P < 0.05) 50 min after 21% estimated blood volume withdrawal. The same rats developed increased tissue hypoxia after hemorrhage, as indicated by prolonged elevations in lactate (2.77 ± 0.5 vs. 1.34 ± 0.2 mmol/l, 60 min after start of hemorrhage, P < 0.05). 5-HT1A mRNA levels in the commissural NTS were directly correlated with renal sympathetic nerve activity (P < 0.01) and inversely correlated with lactate (P < 0.05) 60 min after start of hemorrhage. The data suggest that 5-HT1A receptors in the commissural NTS facilitate tissue perfusion after blood loss likely by increasing sympathetic-mediated venous return.


Subject(s)
Baroreflex , Hemorrhage/metabolism , Hypotension/metabolism , Kidney/innervation , Receptor, Serotonin, 5-HT1A/metabolism , Serotonergic Neurons/metabolism , Solitary Nucleus/metabolism , Sympathetic Nervous System/metabolism , Animals , Baroreflex/drug effects , Blood Pressure , Blood Volume , Disease Models, Animal , Hemorrhage/genetics , Hemorrhage/physiopathology , Hypotension/genetics , Hypotension/physiopathology , Lactic Acid/metabolism , Male , RNA Interference , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/drug effects , Receptor, Serotonin, 5-HT1A/genetics , Recovery of Function , Serotonergic Neurons/drug effects , Serotonin 5-HT1 Receptor Agonists/pharmacology , Signal Transduction , Solitary Nucleus/drug effects , Solitary Nucleus/physiopathology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiopathology , Time Factors
2.
Horm Behav ; 64(2): 203-10, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23998664

ABSTRACT

This article is part of a Special Issue "Puberty and Adolescence". Sexual differentiation is the process by which the nervous system becomes structurally and functionally dissimilar in females and males. In mammals, this process has been thought to occur during prenatal and early postnatal development, when a transient increase in testosterone secretion masculinizes and defeminizes the developing male nervous system. Decades of research have led to the views that structural sexual dimorphisms created during perinatal development are passively maintained throughout life, and that ovarian hormones do not play an active role in feminization of the nervous system. Furthermore, perinatal testosterone was thought to determine sex differences in neuron number by regulating cell death and cell survival, and not by regulating cell proliferation. As investigations of neural development during adolescence became more prominent in the late 20th century and revealed the extent of brain remodeling during this time, each of these tenets has been challenged and modified. Here we review evidence from the animal literature that 1) the brain is further sexually differentiated during puberty and adolescence; 2) ovarian hormones play an active role in the feminization of the brain during puberty; and 3) hormonally modulated, sex-specific addition of new neurons and glial cells, as well as loss of neurons, contribute to sexual differentiation of hypothalamic, limbic, and cortical regions during adolescence. This architectural remodeling during the adolescent phase of sexual differentiation of the brain may underlie the known sex differences in vulnerability to addiction and psychiatric disorders that emerge during this developmental period.


Subject(s)
Brain/growth & development , Hormones/physiology , Rodentia/physiology , Sex Differentiation/physiology , Sexual Maturation/physiology , Animals , Female , Humans , Hypothalamus/growth & development , Male
3.
Dev Cogn Neurosci ; 57: 101141, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35933923

ABSTRACT

The rodent posterodorsal medial amygdala (MePD) evaluates and assigns valence to social sensory stimuli. The perception of social stimuli evolves during puberty, when the focus of social interactions shifts from kin to peers. Using the cell birthdate marker bromo-deoxyuridine (BrdU), we previously discovered that more pubertally born cells are added to the rat MePD in males than females. Here we addressed several questions that remained unanswered by our previous work. First, to determine whether there are sex differences in cell proliferation within the MePD, we examined BrdU-immunoreactive (-ir) cells at 2 and 4 h following BrdU administration on postnatal day 30 (P30). The density of BrdU-ir cells was greater in males than in females, indicating greater proliferation in males. Proliferation was substantiated by double-label immunohistochemistry showing that MePD BrdU-ir cells colocalize proliferating cell nuclear antigen, but not the cell death marker Caspase3. We next studied longer time points (2-21 days) following BrdU administration on P30 and found that the rate of cell attrition is higher in males. Finally, triple-label immunohistochemistry of P30-born MePD cells revealed that some of these cells differentiate into neurons or astrocytes within three weeks of cell birth, with no discernable sex differences. The demonstration of pubertal neuro- and glio-genesis in the MePD of male and female rats adds a new dimension to developmental plasticity of the MePD that may contribute to pubertal changes in the perception of social stimuli in both sexes.

4.
Reproduction ; 135(4): 419-29, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18367504

ABSTRACT

Neuron-to-glia, glia-to-neuron, and glia-to-glia communication are implicated in the modulation of neuronal activity and synaptic transmission relevant to reproduction. Glial cells play an important role in neuroendocrine regulation and participate in the sexual differentiation of neuronal connectivity of brain regions involved in the control of reproductive neuroendocrine output. During puberty, modifications in the morphology and chemistry of astrocytes and tanycytes in the hypothalamus and median eminence influence the maturation of the neuronal circuits controlling the secretion of GnRH. During adult reproductive life, the glial cells participate in the transient remodeling of neuronal connectivity in the preoptic area, the arcuate nucleus, the median eminence, and other brain regions involved in the control of reproduction. Gonadal hormones regulate glial plasticity by direct and indirect effects and regulate various other endocrine signals, local soluble factors and adhesion molecules that also affect glial function and glia-to-neuron communication. The glial cells, therefore, are central to the coordination of endocrine and local inputs that bring about neural plasticity and adapt reproductive capacity to homeostatic signals.


Subject(s)
Gonadotropin-Releasing Hormone/physiology , Hypothalamus , Neuroglia/physiology , Neurons/physiology , Neurosecretory Systems/physiology , Reproduction/physiology , Adult , Cell Communication , Humans , Neuronal Plasticity
5.
eNeuro ; 4(5)2017.
Article in English | MEDLINE | ID: mdl-29098175

ABSTRACT

New cells are added during both puberty and adulthood to hypothalamic regions that govern reproduction, homeostasis, and social behaviors, yet the functions of these late-born cells remain elusive. Here, we pharmacologically inhibited cell proliferation in ventricular zones during puberty or in adulthood and determined subsequent effects on the hormone-induced surge of luteinizing hormone (LH) in female rats. Initial neuroanatomical analyses focused on verifying incorporation, activation, and pharmacological inhibition of pubertally or adult born cells in the anteroventral periventricular nucleus (AVPV) of the hypothalamus because of the essential role of the AVPV in triggering the preovulatory LH surge in females. We first showed that approximately half of the pubertally born AVPV cells are activated by estradiol plus progesterone (P) treatment, as demonstrated by Fos expression, and that approximately 10% of pubertally born AVPV cells express estrogen receptor alpha (ERα). Next, we found that mitotic inhibition through intracerebroventricular (ICV) administration of cytosine ß-D-arabinofuranoside (AraC), whether during puberty or in adulthood, decreased the number of new cells added to the AVPV and the suprachiasmatic nucleus (SCN), and also blunted and delayed the hormone-induced LH surge. These studies do not prove, but are highly suggestive, that ongoing postnatal addition of new cells in periventricular brain regions, including the AVPV and SCN, may be important to the integrity of female reproduction.


Subject(s)
Hypothalamus, Anterior/cytology , Hypothalamus, Anterior/metabolism , Luteinizing Hormone/metabolism , Sexual Maturation/physiology , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/metabolism , Animals , Antimitotic Agents/pharmacology , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Estradiol/administration & dosage , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Female , Hypothalamus, Anterior/drug effects , Hypothalamus, Anterior/growth & development , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Ovary/growth & development , Ovary/metabolism , Progesterone/administration & dosage , Progesterone/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Receptors, Progesterone/metabolism , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/growth & development
6.
J Neurosci ; 25(16): 4004-13, 2005 Apr 20.
Article in English | MEDLINE | ID: mdl-15843602

ABSTRACT

In the hamster facial nerve injury paradigm, we have established that androgens enhance both functional recovery from facial nerve paralysis and the rate of regeneration in the adult, through intrinsic effects on the nerve cell body response to injury and via an androgen receptor (AR)-mediated mechanism. Whether these therapeutic effects of gonadal steroids encompass neuroprotection from axotomy-induced cell death is the focus of the present study. Virtually 100% of adult hamster facial motoneurons (FMNs) survive axotomy at the stylomastoid foramen (SMF), whereas, before postnatal day 15 (P15), developing FMNs undergo substantial axotomy-induced cell death. The first part of the present study focuses on determining when ARs are first expressed in developing hamster FMNs. Using AR immunocytochemistry, it was found that males express ARs by P2 and females by P4, which is the earliest demonstration of AR expression in mammalian motoneurons reported thus far in the literature. The second half examines the neuroprotective effects of testosterone propionate, 17-beta estradiol, and dihydrotestosterone on FMNs of P7 hamsters after facial nerve transection at the SMF. The results demonstrate that androgens and estrogens are equally able to rescue approximately 20% of FMNs from axotomy-induced cell death, with the effects permanent. This study is the first to investigate the effects of both androgens and estrogens on axotomy-induced cell death in one system and, with our previously published work, to validate the hamster FMN injury paradigm as a model of choice in the investigation of both neurotherapeutic and neuroprotective actions of gonadal steroids.


Subject(s)
Facial Nerve Injuries/drug therapy , Facial Nerve Injuries/pathology , Gene Expression Regulation, Developmental/drug effects , Gonadal Steroid Hormones/pharmacology , Motor Neurons/drug effects , Age Factors , Animals , Animals, Newborn , Axotomy/methods , Brain Stem/drug effects , Brain Stem/growth & development , Brain Stem/pathology , Cell Count/methods , Cell Death/drug effects , Cricetinae , Dihydrotestosterone/pharmacology , Dihydrotestosterone/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Estradiol/pharmacology , Estradiol/therapeutic use , Female , Functional Laterality , Gene Expression Regulation, Developmental/physiology , Gonadal Steroid Hormones/therapeutic use , Immunohistochemistry/methods , Male , Mesocricetus , Motor Neurons/pathology , Receptors, Androgen/immunology , Receptors, Androgen/metabolism , Sex Factors , Testosterone/pharmacology , Testosterone/therapeutic use
7.
Endocrinology ; 157(6): 2393-402, 2016 06.
Article in English | MEDLINE | ID: mdl-27145006

ABSTRACT

The anteroventral periventricular nucleus (AVPV) orchestrates the neuroendocrine-positive feedback response that triggers ovulation in female rodents. The AVPV is larger and more cell-dense in females than in males, and during puberty, only females develop the capacity to show a positive feedback response. We previously reported a potential new mechanism to explain this female-specific gain of function during puberty, namely a female-biased sex difference in the pubertal addition of new cells to the rat AVPV. Here we first asked whether this sex difference is due to greater cell proliferation and/or survival in females. Female and male rats received the cell birthdate marker 5-bromo-2'-deoxyuridine (BrdU; 200 mg/kg, ip) on postnatal day (P) 30; brains were collected at short and long intervals after BrdU administration to assess cell proliferation and survival, respectively. Overall, females had more BrdU-immunoreactive cells in the AVPV than did males, with no sex differences in the rate of cell attrition over time. Thus, the sex difference in pubertal addition of AVPV cells appears to be due to greater cell proliferation in females. Next, to determine the phenotype of pubertally born AVPV cells, daily BrdU injections were given to female rats on P28-56, and tissue was collected on P77 to assess colocalization of BrdU and markers for mature neurons or glia. Of the pubertally born AVPV cells, approximately 15% differentiated into neurons, approximately 19% into astrocytes, and approximately 23% into microglia. Thus, both neuro- and gliogenesis occur in the pubertal female rat AVPV and potentially contribute to maturation of female reproductive function.


Subject(s)
Hypothalamus, Anterior/cytology , Hypothalamus, Anterior/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cell Proliferation/physiology , Cell Survival/physiology , Female , Male , Microglia/cytology , Microglia/metabolism , Puberty/physiology , Rats , Rats, Sprague-Dawley , Sexual Maturation/physiology
8.
Endocrinology ; 146(10): 4340-8, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16002522

ABSTRACT

Reproductive and behavioral functions of progesterone receptors (PRs) in males were assessed by examining consequences of PR gene deletion. Basal hormone levels were measured in male progesterone receptor knockout (PRKO) mice and compared to wild-type (WT) counterparts. RIA of serum LH, testosterone, and progesterone levels revealed no significant differences. Levels of FSH were moderately but significantly lower and inhibin levels were higher in PRKOs; these differences were not accompanied by gross differences in testicular weight or morphology. PRKOs exhibited significant alterations in sexual behavior. In initial tests PRKOs exhibited reduced latency to mount, compared with WT. In second sessions, PRKOs again showed a significantly reduced latency to mount and increased likelihood of achieving ejaculation. RU486 treatment in WT produced increased mount and intromission frequency and decreased latency to intromission. In anxiety-related behavior tests, PRKO mice exhibited intermediate anxiety levels, compared with WT, suggesting that enhanced sexual behavior in PRKOs is not secondary to reduced anxiety. Immunohistochemical analysis revealed significantly enhanced androgen receptor expression in the medial preoptic nucleus and bed nucleus of the stria terminalis of PRKO. We conclude that testicular development and function and homeostatic regulation of the hypothalamic-pituitary testicular axis are altered to a lesser extent by PR gene deletion. In contrast, PR appears to play a substantial role in inhibiting the anticipatory/motivational components of male sexual behavior in the mouse. The biological significance of this inhibitory mechanism and the extent to which it is mediated by reduced androgen receptor expression remain to be clarified.


Subject(s)
Receptors, Androgen/physiology , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics , Sexual Behavior, Animal , Animals , Anxiety , DNA Primers , Follicle Stimulating Hormone/blood , Luteinizing Hormone/blood , Male , Mice , Mice, Knockout , Mifepristone/pharmacology , Motor Activity , Organ Size , Polymerase Chain Reaction , Progesterone/pharmacology , Radioimmunoassay , Sperm Count , Testis/anatomy & histology
9.
J Comp Neurol ; 492(4): 456-68, 2005 Nov 28.
Article in English | MEDLINE | ID: mdl-16228996

ABSTRACT

Androgen exposure during development and adulthood promotes cell-to-cell communication, modulates the size of specific brain nuclei, and influences hormone-dependent behavioral and neuroendocrine functions. Androgen action involves the activation of androgen receptors (AR). To elucidate the mechanisms involved in AR-mediated effects on forebrain development, double-label fluorescent immunohistochemistry and confocal microscopy were employed to identify the cellular phenotype of AR-immunoreactive (AR(+)) cells in the developing (embryonic day 20, postnatal days 0, 4, 10) and adult male rat forebrain. Sections were doubly labeled with antibodies directed against AR and one of the following: neurons (immature, nestin; mature, NeuN) or astrocytes [immature, vimentin; mature, glial fibrillary acidic protein (GFAP)] or mature oligodendrocytes (mGalC). In all brain regions examined, by far the majority of AR(+) cells were neurons. In addition, small subsets of AR(+) cells were identified as mature astrocytes (GFAP(+)) but only in specific brain regions at specific ages. AR(+)/GFAP(+) cells were observed in the cerebral cortex but only in postnatal day 10 rats and in the arcuate nucleus of the hypothalamus but only in adult rats. Immature neurons, immature astrocytes, and oligodendrocytes were not AR(+) at any age, in any region. Thus, both neurons and astrocytes in the male rat forebrain contain ARs, suggesting that androgens, via ARs, may exert effects on both cell types in an age- and region-dependent manner.


Subject(s)
Brain/anatomy & histology , Brain/metabolism , Receptors, Androgen/metabolism , Androgens/pharmacology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Biomarkers/metabolism , Brain/drug effects , Brain/growth & development , Female , Immunohistochemistry , Male , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Phenotype , Rats , Rats, Sprague-Dawley
10.
Aging Cell ; 2(1): 31-7, 2003 02.
Article in English | MEDLINE | ID: mdl-12882332

ABSTRACT

Human aging is associated with a decrease of circulating gonadal steroid hormones. Since these hormones act as trophic factors for neurones and glia, it is possible that the decrease in sex steroid levels may contribute to the increased risk of neurodegenerative disorders with advanced age. Sex steroids are neuroprotective in several animal models of central and peripheral neurodegenerative diseases, and clinical data suggest that these hormones may reduce the risk of neural pathology in aged humans. Potential therapeutic approaches for aged-associated neural disorders may emerge from studies conducted to understand the mechanisms of action of sex steroids in the nervous system of aged animals. Alterations in the endogenous capacity of the aged brain to synthesize and metabolize sex steroids, as well as possible aged-associated modifications in the signalling of sex steroid receptors in the nervous system, are important areas for future investigation.


Subject(s)
Aging/physiology , Brain/physiology , Gonadal Steroid Hormones/metabolism , Animals , Gonadal Steroid Hormones/therapeutic use , Hormone Replacement Therapy , Humans , Neuroprotective Agents/metabolism
11.
Sleep Health ; 1(4): 233-243, 2015 Dec.
Article in English | MEDLINE | ID: mdl-29073398

ABSTRACT

OBJECTIVE: To make scientifically sound and practical recommendations for daily sleep duration across the life span. METHODS: The National Sleep Foundation convened a multidisciplinary expert panel (Panel) with broad representation from leading stakeholder organizations. The Panel evaluated the latest scientific evidence and participated in a formal consensus and voting process. Then, the RAND/UCLA Appropriateness Method was used to formulate sleep duration recommendations. RESULTS: The Panel made sleep duration recommendations for 9 age groups. Sleep duration ranges, expressed as hours of sleep per day, were designated as recommended, may be appropriate, or not recommended. Recommended sleep durations are as follows: 14-17 hours for newborns, 12-15 hours for infants, 11-14 hours for toddlers, 10-13 hours for preschoolers, 9-11 hours for school-aged children, and 8-10 hours for teenagers. Seven to 9 hours is recommended for young adults and adults, and 7-8 hours of sleep is recommended for older adults. The self-designated basis for duration selection and critical discussions are also provided. CONCLUSIONS: Consensus for sleep duration recommendations was reached for specific age groupings. Consensus using a multidisciplinary expert Panel lends robust credibility to the results. Finally, limitations and caveats of these recommendations are discussed.

12.
Sleep Health ; 1(1): 40-43, 2015 Mar.
Article in English | MEDLINE | ID: mdl-29073412

ABSTRACT

OBJECTIVE: The objective was to conduct a scientifically rigorous update to the National Sleep Foundation's sleep duration recommendations. METHODS: The National Sleep Foundation convened an 18-member multidisciplinary expert panel, representing 12 stakeholder organizations, to evaluate scientific literature concerning sleep duration recommendations. We determined expert recommendations for sufficient sleep durations across the lifespan using the RAND/UCLA Appropriateness Method. RESULTS: The panel agreed that, for healthy individuals with normal sleep, the appropriate sleep duration for newborns is between 14 and 17 hours, infants between 12 and 15 hours, toddlers between 11 and 14 hours, preschoolers between 10 and 13 hours, and school-aged children between 9 and 11 hours. For teenagers, 8 to 10 hours was considered appropriate, 7 to 9 hours for young adults and adults, and 7 to 8 hours of sleep for older adults. CONCLUSIONS: Sufficient sleep duration requirements vary across the lifespan and from person to person. The recommendations reported here represent guidelines for healthy individuals and those not suffering from a sleep disorder. Sleep durations outside the recommended range may be appropriate, but deviating far from the normal range is rare. Individuals who habitually sleep outside the normal range may be exhibiting signs or symptoms of serious health problems or, if done volitionally, may be compromising their health and well-being.

13.
Endocrinology ; 144(8): 3632-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12865346

ABSTRACT

As members of the steroid receptor superfamily, androgen receptors (ARs) have been traditionally identified as transcription factors. In the presence of ligand, ARs reside in the nucleus, where, upon ligand binding, the receptors dimerize and bind to specific response elements in the promoter region of hormone-responsive genes. However, in this report, we describe the discovery that ARs are also present in axons and dendrites within the mammalian central nervous system. AR expression in axons was identified in the rat brain at the light microscopic level using two different antibodies directed against the N terminus of the AR protein and nickel intensified 3'-3'-diaminobenzidine, and also using fluorescence methods and confocal microscopy. This distribution was confirmed at the ultrastructural level. In addition, AR immunoreactivity was identified in small dendrites at the ultrastructural level. AR-immunoreactive axons were observed primarily in the cerebral cortex and were rare in regions where nuclear AR expression is abundant. The observation that ARs are present in axons and dendrites highlights the possibility that androgens play an important and novel extra-nuclear role in neuronal function.


Subject(s)
Axons/chemistry , Dendrites/chemistry , Prosencephalon/ultrastructure , Receptors, Androgen/analysis , Amygdala/ultrastructure , Animals , Cell Nucleus/chemistry , Cerebral Cortex/ultrastructure , Fluorescent Antibody Technique , Hypothalamus/ultrastructure , Immunohistochemistry , Male , Microscopy, Confocal , Preoptic Area/ultrastructure , Prosencephalon/chemistry , Rats , Rats, Wistar , Septal Nuclei/ultrastructure , Tissue Distribution
14.
J Comp Neurol ; 450(3): 256-71, 2002 Aug 26.
Article in English | MEDLINE | ID: mdl-12209854

ABSTRACT

Estrogens and androgens can protect neurons from death caused by injury to the central nervous system. Astrocytes and microglia are major players in events triggered by neural lesions. To determine whether glia are direct targets of estrogens or androgens after neural insults, steroid receptor expression in glial cells was assessed in two different lesion models. An excitotoxic injury to the hippocampus or a stab wound to the parietal cortex and hippocampus was performed in male rats, and the resultant expression of steroid receptors in glial cells was assessed using double-label immunohistochemistry. Both lesions induced the expression of estrogen receptors (ERs) and androgen receptors (ARs) in glial cells. ERalpha was expressed in astrocytes immunoreactive (ERalpha-ir) for glial fibrillary acidic protein or vimentin. AR immunoreactivity colocalized with microglial markers, such as Griffonia simplicifolia lectin-1 or OX-6. The time course of ER and AR expression in glia was studied in the stab wound model. ERalpha-ir astrocytes and AR-ir microglia were observed 3 days after lesion. The number of ERalpha-ir and AR-ir glial cells reached a maximum 7 days after lesion and returned to low levels by 28 days postinjury. The studies of ERbeta expression in glia were inconclusive; different results were obtained with different antibodies. In sum, these results suggest that reactive astrocytes and reactive microglia are a direct target for estrogens and androgens, respectively.


Subject(s)
Brain Injuries/metabolism , Nerve Degeneration/metabolism , Nerve Regeneration/physiology , Neuroglia/metabolism , Plant Lectins , Rats, Wistar/growth & development , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Brain/physiopathology , Brain Injuries/pathology , Brain Injuries/physiopathology , Disease Models, Animal , Estrogen Receptor alpha , Estrogen Receptor beta , Glial Fibrillary Acidic Protein/metabolism , Gliosis/metabolism , Gliosis/pathology , Gliosis/physiopathology , Immunohistochemistry , Kainic Acid , Lectins/metabolism , Male , Microglia/cytology , Microglia/metabolism , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Rats , Rats, Wistar/injuries , Rats, Wistar/metabolism , Vimentin/metabolism
15.
Exp Gerontol ; 39(11-12): 1623-31, 2004.
Article in English | MEDLINE | ID: mdl-15582278

ABSTRACT

Sex steroids exert pleiotropic effects in the nervous system, preserving neural function and promoting neuronal survival. Therefore, the age-related decrease in sex steroids may have a negative impact on neural function. Progesterone, testosterone and estradiol prevent neuronal loss in the central nervous system in different experimental animal models of neurodegeneration. Furthermore, progesterone and its reduced derivatives dihydroprogesterone and tetrahydroprogesterone reduce aging-associated morphological abnormalities of myelin and aging-associated myelin fiber loss in rat peripheral nerves. However, the results from hormone replacement studies in humans are thus far inconclusive. A possible alternative to hormonal replacement therapy is to increase local steroidogenesis by neural tissues, which express enzymes for steroid synthesis and metabolism. Proteins involved in the intramitochondrial trafficking of cholesterol, the first step in steroidogenesis, such as the peripheral-type benzodiazepine receptor and the steroidogenic acute regulatory protein, are up-regulated in the nervous system after injury. Furthermore, steroidogenic acute regulatory protein expression is increased in the brain of 24-month-old rats compared with young adult rats. This suggests that brain steroidogenesis may be modified in adaptation to neurodegenerative conditions and to the brain aging process. Furthermore, recent studies have shown that local formation of estradiol in the brain, by the enzyme aromatase, is neuroprotective. Therefore, steroidogenic acute regulatory protein, peripheral-type benzodiazepine receptor and aromatase are attractive pharmacological targets to promote neuroprotection in the aged brain.


Subject(s)
Aging/physiology , Brain/physiology , Gonadal Steroid Hormones/physiology , Aged , Animals , Aromatase/metabolism , Brain/metabolism , Female , Hormone Replacement Therapy , Humans , Male , Models, Animal , Neurodegenerative Diseases/metabolism , Phosphoproteins/metabolism , Progesterone/metabolism , Rats , Receptors, GABA-A/metabolism
16.
J Steroid Biochem Mol Biol ; 83(1-5): 211-7, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12650718

ABSTRACT

In the brain, as in other tissues, estradiol interacts with growth factors. One of the growth factors that is involved in the neural actions of estradiol is insulin-like growth factor-I (IGF-I). Estradiol and IGF-I cooperate in the central nervous system to regulate neuronal development, neural plasticity, neuroendocrine events and the response of neural tissue to injury. The precise molecular mechanisms involved in these interactions are still not well understood. In the central nervous system there is abundant co-expression of estrogen receptors (ERs) and IGF-I receptors (IGF-IRs) in the same cells. Furthermore, the expression of estrogen receptors and IGF-I receptors in the brain is cross-regulated. In addition, using specific antibodies for the phosphorylated forms of extracellular-signal regulated kinase (ERK) 1 and ERK2 and Akt/protein kinase B (Akt/PKB) it has been shown that estradiol affects IGF-I signaling pathways in the brain. Estradiol treatment results in a dose-dependent increase in the phosphorylation of ERK and Akt/PKB in the brain of adult ovariectomized rats. In addition, estradiol and IGF-I have a synergistic effects on the activation of Akt/PKB in the adult rat brain. These findings suggest that estrogen effects in the brain may be mediated in part by the activation of the signaling pathways of the IGF-I receptor.


Subject(s)
Brain/metabolism , Estrogens/metabolism , Insulin-Like Growth Factor I/metabolism , Animals , Cell Differentiation , Central Nervous System/metabolism , Enzyme Activation , Gonadotropins/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Protein Binding , Receptors, Estrogen/metabolism , Signal Transduction , Time Factors
17.
Dev Neurobiol ; 74(6): 633-42, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24339170

ABSTRACT

Adolescence involves shifts in social behaviors, behavioral flexibility, and adaptive risk-taking that coincide with structural remodeling of the brain. We previously showed that new cells are added to brain regions associated with sexual behaviors, suggesting that cytogenesis may be a mechanism for acquiring adult-typical behaviors during adolescence. Whether pubertal cell addition occurs in brain regions associated with behavioral flexibility or motivation and whether these patterns differ between pubertal and adult animals had not been determined. Therefore, we assessed patterns of cell proliferation or survival in the prefrontal cortex and nucleus accumbens. Pubertal and adult male rats were given injections of bromo-deoxyuridine (BrdU). To assess cell proliferation, half of the animals from each group were sacrificed 24 h following the last injection. The remaining animals were sacrificed at Day 30 following the last injection to evaluate cell survival. Adult animals had significantly lower densities of BrdU-immunoreactive (ir) cells in the prefrontal cortex, irrespective of post-BrdU survival time, whereas in the nucleus accumbens, adult animals had a lower density of BrdU-ir cells at the short survival time; however, the density of BrdU-ir cells was equivalent in pubertal and adult animals at the longer survival time. These data provide evidence that cell addition during puberty may contribute to the remodeling of brain regions associated with behavioral flexibility and motivation, and this cell addition continues into adulthood, albeit at lower levels. Higher levels of cell proliferation or survival in younger animals may reflect a higher level of plasticity, possibly contributing to the dynamic remodeling of the pubertal brain.


Subject(s)
Aging , Neurogenesis/physiology , Neurons/physiology , Nucleus Accumbens/cytology , Nucleus Accumbens/growth & development , Prefrontal Cortex/cytology , Prefrontal Cortex/growth & development , Analysis of Variance , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Male , Rats , Rats, Sprague-Dawley , Time Factors
18.
J Comp Neurol ; 520(11): 2531-44, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22581688

ABSTRACT

The posterodorsal medial amygdala (MePD) exhibits numerous sex differences including differences in volume and in the number and morphology of neurons and astroctyes. In adulthood, gonadal hormones, including both androgens and estrogens, have been shown to play a role in maintaining the masculine character of many of these sex differences, but whether adult gonadal hormones maintain the increased number and complexity of astrocytes in the male MePD was unknown. To answer this question we examined astrocytes in the MePD of male and female Long Evans rats that were gonadectomized as adults and treated for 30 days with either testosterone or a control treatment. At the end of treatment brains were collected and immunostained for glial fibrillary acidic protein. Stereological analysis revealed that adult androgen levels influenced the number and complexity of astrocytes in the MePD of both sexes, but the specific effects of androgens were different in males and females. However, sex differences in the number and complexity of adult astrocytes persisted even in the absence of gonadal hormones in adulthood, suggesting that androgens also act earlier in life to determine these adult sex differences. Using immunofluorescence and confocal microscopy, we found robust androgen receptor immunostaining in a subpopulation of MePD astrocytes, suggesting that testosterone may act directly on MePD astrocytes to influence their structure and function.


Subject(s)
Amygdala/physiology , Astrocytes/physiology , Testosterone/physiology , Amygdala/cytology , Animals , Cell Count , Female , Glial Fibrillary Acidic Protein/metabolism , Male , Random Allocation , Rats , Rats, Long-Evans , Sex Characteristics
19.
Horm Mol Biol Clin Investig ; 1(1): 21-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-25961968

ABSTRACT

The nervous system is a steroidogenic tissue and several steroids synthesized locally in the brain, such as pregnenolone, progesterone and estradiol, modulate neuronal and glial physiology and are neuroprotective. The brain upregulates steroidogenesis at sites of injury as part of a program triggered by neural tissue to cope with neurodegenerative insults. Pharmacological targets to increase brain steroidogenesis and promote neuroprotection include the molecules that transport cholesterol to the inner mitochondrial membrane, where the first enzyme for steroidogenesis is located. Furthermore, the human gene encoding aromatase, the enzyme that synthesizes estradiol, is under the control of different tissue-specific promoters, and it is therefore conceivable that selective aromatase modulators can be developed that will enhance the expression of the enzyme and the consequent increase in estrogen formation in the brain but not in other tissues.

20.
Psychoneuroendocrinology ; 35(7): 1023-33, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20138435

ABSTRACT

Estradiol regulates serotonin 1A (5-HT(1A)) receptor signaling. Since desensitization of 5-HT(1A) receptors may be an underlying mechanism by which selective serotonin reuptake inhibitors (SSRIs) mediate their therapeutic effects and combining estradiol with SSRIs enhances the efficacy of the SSRIs, it is important to determine which estrogen receptors are capable of desensitizating 5-HT(1A) receptor function. We previously demonstrated that selective activation of the estrogen receptor, GPR30, desensitizes 5-HT(1A) receptor signaling in rat hypothalamic paraventricular nucleus (PVN). However, since estrogen receptor-beta (ERbeta), is highly expressed in the PVN, we investigated the role of ERbeta in estradiol-induced desensitization of 5-HT(1A) receptor signaling. We first showed that a selective ERbeta agonist, diarylpropionitrile (DPN) has a 100-fold lower binding affinity than estradiol for GPR30. Administration of DPN did not desensitize 5-HT(1A) receptor signaling in rat PVN as demonstrated by agonist-stimulated hormone release. Second, we used a recombinant adenovirus containing ERbeta siRNAs to decrease ERbeta expression in the PVN. Reductions in ERbeta did not alter the estradiol-induced desensitization of 5-HT(1A) receptor signaling in oxytocin cells. In contrast, in animals with reduced ERbeta, estradiol administration, instead of producing desensitization, augmented the ACTH response to a 5-HT(1A) agonist. Combined with the results from the DPN treatment experiments, desensitization of 5-HT(1A) receptor signaling does not appear to be mediated by ERbeta in oxytocin cells, but that ERbeta, together with GPR30, may play a complex role in central regulation of 5-HT(1A)-mediated ACTH release. Determining the mechanisms by which estrogens induce desensitization may aid in the development of better treatments for mood disorders.


Subject(s)
Estradiol/metabolism , Estrogen Receptor beta/drug effects , Estrogen Receptor beta/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/metabolism , Animals , Estradiol/pharmacology , Female , Nitriles/pharmacology , Oxytocin/blood , Oxytocin/metabolism , Propionates/pharmacology , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL