1
|
Cory-Slechta DA, Downs CJ, Sobolewski M. Cumulative risk assessment as the pathway to public health protection for behavioral neurotoxicity. Neurotoxicology 2025; 108:400-411. [PMID: 40349850 DOI: 10.1016/j.neuro.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/09/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
The formulation of adverse outcome pathways (AOPs) based on high-throughput in vitro new approach methods linking biochemical/mechanistic data with an apical endpoint considered an adverse outcome (AO), is increasingly proposed to accelerate the process of risk assessment for environmental chemical exposures. While a laudable goal, this approach ignores the extensive evidence demonstrating context-dependence of neurotoxicological consequences, including behavioral toxicity of chemical exposures. Such contextual modifiers can include environmental conditions (poverty, psychosocial stress, behavioral experience/history), physiological conditions (sex, period of exposure, nutritional status, brain region, exposure parameters), and genetic background. Context dependence represents a serious omission for AOP formulation because an environmental context can alter a chemical's molecular targets, or potentially enhance toxicity through interactions with other contextual conditions, thus leading to potential underestimation of neurological risks due to such exposures. The integrative physiological basis of AOs requires cumulative risk assessments that model environmental contexts across scales of biology, i.e., integration and testing in whole-animal models. AOPs contribute to the derivation of cumulative risk considerations regarding factors to incorporate into cumulative risk assessments by defining risk factors with shared biological targets. Epidemiological and animal model studies can provide information to prioritize interactive effects of greatest magnitude. Additionally, a focus on how a single risk factor in different physiological contexts may attribute risk across multiple neurologic conditions, rather than to a single unique condition, would provide broader public health protection. Realistic acknowledgement of context-dependence is requisite to understanding both the etiological basis of neurological diseases and disorders and to human health protection.
Collapse
Affiliation(s)
- Deborah A Cory-Slechta
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States.
| | - Cynthia J Downs
- Department of Environmental Biology, SUNY College of Environmental Science and Forestry, Syracuse, NY 13210, United States
| | - Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States.
| |
Collapse
|
2
|
Pizaña-Encarnación JM, Escoto-Rosales MJ, Islas-Espinoza AM, Morales-Galindo DK, Déciga-Campos M, Quintanar BG, Reyes R, Granados-Soto V, Fernández-Guasti A. Activational and organizational actions of gonadal hormones on the sexual dimorphism of the α 6-subunit containing GABA A receptor in Wistar rats with neuropathic pain. Horm Behav 2025; 171:105746. [PMID: 40250165 DOI: 10.1016/j.yhbeh.2025.105746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/21/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Sex differences in pain perception and response to analgesics are well documented, yet the underlying causes remain poorly understood. Here we investigate the sexual dimorphism in the function of α6GABAA receptors in neuropathic pain, focusing on activational and organizational actions of gonadal hormones. Using the nerve ligation model in rats, we found that the positive allosteric modulator, PZ-ll-029 (30 nmol, it), produced a robust antiallodynic effect in females but not in males. Ovariectomy abolished this effect, while a single dose of estradiol (20 μg/kg sc, -24 h), that returned to physiological serum levels, partially restored it, indicating that the activational effect of estradiol is crucial for α6GABAA receptor-mediated antiallodynia in females. Interestingly, adult or neonatal (at postnatal day 3) orchidectomy did not alter the male's insensitivity to PZ-ll-029, even after estradiol treatment. However, neonatal female's virilization (with testosterone propionate 120 μg/rat at postnatal day 3) induced a male-like insensitivity to PZ-ll-029, that was partial when the ovaries were present and complete after adult ovariectomy. These findings reveal that the neonatal organizational effects of testosterone determine the sex-specific insensitivity of α6GABAA receptors to modulate neuropathic pain, while the activational effects of estradiol can partly maintain the female-typical response, despite early androgen exposure. Our results provide new insights into hormonal regulation of pain modulation and suggest that both developmental exposure and adult status should be considered in basic research and preclinical studies investigating sex-based dimorphisms.
Collapse
Affiliation(s)
| | - María José Escoto-Rosales
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Ana M Islas-Espinoza
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Diana Karen Morales-Galindo
- Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional. Mexico City, Mexico
| | - Myrna Déciga-Campos
- Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional. Mexico City, Mexico
| | | | - Rebeca Reyes
- Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico.
| | | |
Collapse
|
3
|
Brill J, Linden DJ. Chronic Aromatase Inhibition Attenuates Synaptic Plasticity in Ovariectomized Mice. eNeuro 2024; 11:ENEURO.0346-24.2024. [PMID: 39592220 PMCID: PMC11594935 DOI: 10.1523/eneuro.0346-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/11/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Brain-derived estrogen (17β-estradiol, E2) is a neuromodulator that plays important roles in neural plasticity and network excitability. Chronic inhibition of estrogen synthesis is used in adjuvant breast cancer therapy for estrogen receptor-positive tumors and may have been associated with cognitive and affective side effects. Here, we have developed a model of adjuvant therapy in female ovariectomized mice in which the E2 biosynthetic enzyme aromatase is inhibited by letrozole (1 mg/kg/day, i.p., for up to 3 weeks), Using two-photon longitudinal in vivo imaging in Thy1-GFP-M mice, we found that spine density in the apical dendrites of neocortical layer 5 pyramidal cells was unaffected by letrozole treatment but spine turnover was reduced. LTP in layer 4 to layer 2/3 synapses in the somatosensory cortex was also reduced in slices from letrozole-treated mice, showing deficits in structural and functional plasticity resulting from aromatase inhibition. Ovariectomized mice performed worse than intact control mice in the novel object recognition test but, surprisingly, letrozole treatment rescued this deficit.
Collapse
Affiliation(s)
- Julia Brill
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21210
| | - David J Linden
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21210
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, Maryland 21210
| |
Collapse
|
4
|
Liang SL, Chen RS. The Glutamine-Glutamate Cycle Contributes to Behavioral Feminization in Female Rats. Neuroendocrinology 2024; 114:1045-1065. [PMID: 39182491 PMCID: PMC11548894 DOI: 10.1159/000541102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
INTRODUCTION In perinatal female rats, the glutamine (Gln)-glutamate cycle (GGC) constitutively supplies Gln to neurons of the ventral lateral ventromedial nucleus of the hypothalamus (vlVMH) to sustain glutamatergic synaptic transmission (GST). In contrast, male pups may use Gln only during periods of elevated neuronal activity. Perinatal disruption of the GGC has sex-specific effects on the GST and morphology of vlVMH neurons during adulthood. Since (vl)VMH neuronal activities regulate mating behavior expression, we hypothesize that maintaining a perinatal intact GGC may be essential for the sexual differentiation of reproductive behaviors. METHODS Using perinatal rats of both sexes, we pharmacologically killed astrocytes or blocked the GGC and supplemented them with exogenous Gln. Mating behavior, an open-field test and protein levels of GGC enzymes were examined during adulthood. RESULTS Killing astrocytes reduced mating behavior expression by 38-48% and 71-72% in male and female rats, respectively. Any blocker targeting the GGC consistently reduced female lordosis behavior by 52-73% and increased glutaminase protein levels in the hypothalamus, but blockers had no effect on the expression of or motivation for copulatory behavior in males. Exogenous Gln supplementation partly rescued the decline in Gln synthetase inhibitor-mediated sex behavior in females. Perinatal interruption of the GGC did not increase induced expression of female sexual behavior in hormone-primed castrated male rats or affect locomotion or anxiety-like behavior in either sex. CONCLUSION The intact GGC is necessary for behavioral feminization in female rats and may play little or no role in behavioral masculinization or defeminization in males.
Collapse
Affiliation(s)
- Shu-Ling Liang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Rou-Shayn Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| |
Collapse
|
5
|
Yarmohammadi-Samani P, Vatanparast J. Sex-specific dendritic morphology of hippocampal pyramidal neurons in the adolescent and young adult rats. Int J Dev Neurosci 2024; 84:47-63. [PMID: 37933732 DOI: 10.1002/jdn.10307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 09/02/2023] [Accepted: 10/16/2023] [Indexed: 11/08/2023] Open
Abstract
CA1 and CA3 pyramidal neurons are the major sources of hippocampal efferents. The structural features of these neurons are presumed to be involved in various normal/abnormal cognitive and emotional outcomes by influencing the pattern of synaptic inputs and neuronal signal processing. Although many studies have described hippocampal structure differences between males and females, these reports mainly focused on gross anatomical features in adult or aged models, and such distinctions on neuronal morphology and dendritic spine density during adolescence, a period of high vulnerability to neurodevelopmental disorders, have received much less attention. In this work, we analyzed dendritic architecture and density of spines in CA1 and CA3 neurons of male and female rats in early adolescence (postnatal day, PND 40) and compared them with those in late adolescence/young adulthood (PND 60). On PND 40, CA1 neurons of male rats showed more Sholl intersections and spine density in apical and basal dendrites compared to those in females. The Sholl intersections in basal dendrites of CA3 neurons were also more in males, whereas the number of apical dendrite intersections was not significantly different between sexes. In male rats, there was a notable decrease in the number of branch and terminal points in the basal dendrite of CA1 neurons of young adults when compared to their sex-matched adolescent rats. On the other hand, CA1 neurons in young adult females also showed more Sholl intersections in apical and basal dendrites compared to adolescent females. Meanwhile, the total cable length, the number of branches, and terminal points of apical dendrites in CA3 neurons also exhibited a significant reduction in young adult male rats compared to their sex-matched adolescents. In young adult rats, both apical and basal dendrites of CA3 neurons in males showed fewer intersections with Sholl circles, but there were no significant differences in dendritic spine density or count estimation between males and females. On the other hand, young adult female rats had more Sholl intersections and dendritic spine count on the basal dendrites of CA3 neurons compared to adolescent females. Although no significant sex- and age-dependent difference in neuronal density was detected in CA1 and CA3 subareas, CA3 pyramidal neurons of both male and female rats showed reduced soma area compared to adolescent rats. Our findings show that the sex differences in the dendritic structure of CA1 and CA3 neurons vary by age and also by the compartments of dendritic arbors. Such variations in the morphology of hippocampal pyramidal neurons may take part as a basis for normal cognitive and affective differences between the sexes, as well as distinct sensitivity to interfering factors and the prevalence of neuropsychological diseases.
Collapse
Affiliation(s)
| | - Jafar Vatanparast
- Department of Biology, School of Science, Shiraz University, Shiraz, Iran
| |
Collapse
|
6
|
Abstract
Rapid advances in the neural control of social behavior highlight the role of interconnected nodes engaged in differential information processing to generate behavior. Many innate social behaviors are essential to reproductive fitness and therefore fundamentally different in males and females. Programming these differences occurs early in development in mammals, following gonadal differentiation and copious androgen production by the fetal testis during a critical period. Early-life programming of social behavior and its adult manifestation are separate but yoked processes, yet how they are linked is unknown. This review seeks to highlight that gap by identifying four core mechanisms (epigenetics, cell death, circuit formation, and adult hormonal modulation) that could connect developmental changes to the adult behaviors of mating and aggression. We further propose that a unique social behavior, adolescent play, bridges the preweaning to the postpubertal brain by engaging the same neural networks underpinning adult reproductive and aggressive behaviors.
Collapse
Affiliation(s)
- Margaret M McCarthy
- Department of Pharmacology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA;
| |
Collapse
|
7
|
Corbett CM, Miller EN, Wannen EE, Rood BD, Chandler DJ, Loweth JA. Cocaine Exposure Increases Excitatory Synaptic Transmission and Intrinsic Excitability in the Basolateral Amygdala in Male and Female Rats and across the Estrous Cycle. Neuroendocrinology 2023; 113:1127-1139. [PMID: 37271140 PMCID: PMC10623393 DOI: 10.1159/000531351] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/10/2023] [Indexed: 06/06/2023]
Abstract
INTRODUCTION Sex and ovarian hormones influence cocaine seeking and relapse vulnerability, but less is known regarding the cellular and synaptic mechanisms contributing to these behavioral sex differences. One factor thought to influence cue-induced seeking behavior following withdrawal is cocaine-induced changes in the spontaneous activity of pyramidal neurons in the basolateral amygdala (BLA). However, the mechanisms underlying these changes, including potential sex or estrous cycle effects, are unknown. METHODS Ex vivo whole-cell patch clamp electrophysiology was conducted to investigate the effects of cocaine exposure, sex, and estrous cycle fluctuations on two properties that can influence spontaneous activity of BLA pyramidal neurons: (1) frequency and amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) and (2) intrinsic excitability. Recordings of BLA pyramidal neurons were conducted in adult male and female rats and across the estrous cycle following 2-4 weeks of withdrawal from extended-access cocaine self-administration (6 h/day for 10 days) or drug-naïve conditions. RESULTS In both sexes, cocaine exposure increased the frequency, but not amplitude, of sEPSCs and neuronal intrinsic excitability. Across the estrous cycle, sEPSC frequency and intrinsic excitability were significantly elevated only in cocaine-exposed females in the estrus stage of the cycle, a stage when cocaine-seeking behavior is known to be enhanced. CONCLUSIONS Here, we identify potential mechanisms underlying cocaine-induced alterations in the spontaneous activity of BLA pyramidal neurons in both sexes along with changes in these properties across the estrous cycle.
Collapse
Affiliation(s)
- Claire M. Corbett
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Emily N.D. Miller
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Erin E. Wannen
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Benjamin D Rood
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Daniel J. Chandler
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Jessica A. Loweth
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
| |
Collapse
|
8
|
Terrin F, Tesoriere A, Plotegher N, Dalla Valle L. Sex and Brain: The Role of Sex Chromosomes and Hormones in Brain Development and Parkinson's Disease. Cells 2023; 12:1486. [PMID: 37296608 PMCID: PMC10252697 DOI: 10.3390/cells12111486] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Sex hormones and genes on the sex chromosomes are not only key factors in the regulation of sexual differentiation and reproduction but they are also deeply involved in brain homeostasis. Their action is crucial for the development of the brain, which presents different characteristics depending on the sex of individuals. The role of these players in the brain is fundamental in the maintenance of brain function during adulthood as well, thus being important also with respect to age-related neurodegenerative diseases. In this review, we explore the role of biological sex in the development of the brain and analyze its impact on the predisposition toward and the progression of neurodegenerative diseases. In particular, we focus on Parkinson's disease, a neurodegenerative disorder that has a higher incidence in the male population. We report how sex hormones and genes encoded by the sex chromosomes could protect from the disease or alternatively predispose toward its development. We finally underline the importance of considering sex when studying brain physiology and pathology in cellular and animal models in order to better understand disease etiology and develop novel tailored therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Nicoletta Plotegher
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.T.); (A.T.)
| | - Luisa Dalla Valle
- Department of Biology, University of Padova, 35131 Padova, Italy; (F.T.); (A.T.)
| |
Collapse
|
9
|
Beeson ALS, Meitzen J. Estrous cycle impacts on dendritic spine plasticity in rat nucleus accumbens core and shell and caudate-putamen. J Comp Neurol 2023; 531:759-774. [PMID: 36756791 PMCID: PMC10994586 DOI: 10.1002/cne.25460] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/22/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023]
Abstract
An important factor that can modulate neuron properties is sex-specific hormone fluctuations, including the human menstrual cycle and rat estrous cycle in adult females. Considering the striatal brain regions, the nucleus accumbens (NAc) core, NAc shell, and caudate-putamen (CPu), the estrous cycle has previously been shown to impact relevant behaviors and disorders, neuromodulator action, and medium spiny neuron (MSN) electrophysiology. Whether the estrous cycle impacts MSN dendritic spine attributes has not yet been examined, even though MSN spines and glutamatergic synapse properties are sensitive to exogenously applied estradiol. Thus, we hypothesized that MSN dendritic spine attributes would differ by estrous cycle phase. To test this hypothesis, brains from adult male rats and female rats in diestrus, proestrus AM, proestrus PM, and estrus were processed for Rapid Golgi-Cox staining. MSN dendritic spine density, size, and type were analyzed in the NAc core, NAc shell, and CPu. Overall spine size differed across estrous cycle phases in female NAc core and NAc shell, and spine length differed across estrous cycle phase in NAc shell and CPu. Consistent with previous work, dendritic spine density was increased in the NAc core compared to the NAc shell and CPu, independent of sex and estrous cycle. Spine attributes in all striatal regions did not differ by sex when estrous cycle was disregarded. These results indicate, for the first time, that estrous cycle phase impacts dendritic spine plasticity in striatal regions, providing a neuroanatomical avenue by which sex-specific hormone fluctuations can impact striatal function and disorders.
Collapse
Affiliation(s)
- Anna LS Beeson
- Department of Biological Sciences, NC State University, Raleigh, USA
- Graduate Program in Biology, NC State University, Raleigh, USA
| | - John Meitzen
- Department of Biological Sciences, NC State University, Raleigh, USA
- Comparative Medicine Institute, NC State University, Raleigh, USA
- Center for Human Health and the Environment, NC State University, Raleigh, USA
| |
Collapse
|
10
|
Liang SL, Liao WL, Chen RS. Perinatal blockade of neuronal glutamine transport sex-differentially alters glutamatergic synaptic transmission and organization of neurons in the ventrolateral ventral media hypothalamus of adult rats. J Neuroendocrinol 2023; 35:e13253. [PMID: 36949648 DOI: 10.1111/jne.13253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/13/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023]
Abstract
Compared to male pups, perinatal female rats rely heavily on neuronal glutamine (Gln) transport for sustaining glutamatergic synaptic release in neurons of the ventrolateral ventral media nucleus of the hypothalamus (vlVMH). VMH mainly regulates female sexual behavior and increases glutamate release of perinatal hypothalamic neurons, permanently enhances dendrite spine numbers and is associated with brain and behavioral defeminization. We hypothesized that perinatal interruption of neuronal Gln transport may alter the glutamatergic synaptic transmission during adulthood. Perinatal rats of both sexes received an intracerebroventricular injection of a neuronal Gln uptake blocker, alpha-(methylamino) isobutyric acid (MeAIB, 5 mM), and were raised until adulthood. Whole-cell voltage-clamp recordings of miniature excitatory postsynaptic currents (mEPSCs) and evoked EPSCs (eEPSCs) of vlVMH neurons in adult rats with the perinatal pretreatment were conducted and neuron morphology was subjected to post hoc examination. Perinatal MeAIB treatment sex-differentially increased mEPSC frequency in males, but decreased mEPSC amplitude and synaptic Glu release in females. The pretreatment sex-differentially decreased eEPSC amplitude in males but increased AMPA/NMDA current ratio in females, and changed the morphology of vlVMH neurons of adult rats to that of the opposite sex. Most alterations in the glutamatergic synaptic transmission resembled the changes occurring during MeAIB acute exposure in perinatal rats of both sexes. We conclude that perinatal blockade of neuronal Gln transport mediates changes via different presynaptic and postsynaptic mechanisms to induce sex-differential alterations of the glutamatergic synaptic transmission and organization of vlVMH neurons in adult rats. These changes may be permanent and associated with brain and behavior feminization and/or defeminization in rats.
Collapse
Affiliation(s)
- Shu-Ling Liang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Wen-Lin Liao
- Institute of Neuroscience, National Chengchi University, Taipei, Taiwan
| | - Rou-Shayn Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| |
Collapse
|
11
|
Kiyokage E, Ichikawa S, Horie S, Hayashi S, Toida K. Effects of estradiol on dopaminergic synapse formation in the mouse olfactory bulb. J Comp Neurol 2023; 531:528-547. [PMID: 36519231 DOI: 10.1002/cne.25441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 10/24/2022] [Accepted: 11/26/2022] [Indexed: 12/23/2022]
Abstract
Olfactory glomeruli are the sites of initial synaptic integration in olfactory information processing. They are surrounded by juxtaglomerular (JG) cells, which include periglomerular, superficial short axon, and external tufted cells. A subpopulation of JG cells expresses the dopamine synthetic enzymes, tyrosine hydroxylase (TH), and aromatic l-amino acid decarboxylase (AADC). TH cells corelease γ-aminobutyric acid (GABA) and their processes extend to multiple glomeruli forming intra- and interglomerular circuits. It is well established that 17β-estradiol (E2) exerts wide ranging effects in the central nervous system. However, participation of E2 in the modulation of neurotransmission and synaptic plasticity of TH cells in olfactory glomeruli is unknown. To address this, we subcutaneously implanted a 60-day release pellet of E2 or placebo into intact male mice and compared glomerular TH, AADC, and vesicular γ-aminobutyric acid transporter (VGAT) immunoreactivity between them. High-voltage electron microscopy (HVEM) and ultra-HVEM using immunogold revealed significantly increased immunoreactive intensity at the cellular level for TH and AADC after E2 treatment and for VGAT in TH cells. These results indicate that E2 may affect the interplay between dopaminergic and GABAergic systems. Moreover, random-section electron microscopy analysis showed a significant increase in the number of symmetrical synapses from TH cell to mitral/tufted cell dendrites after E2 treatment. This result was supported by quantitative immunofluorescence staining with synapse markers. Together, these data indicate that E2 may regulate inhibition between TH cells and olfactory bulb neurons within the glomerulus via interaction between dopaminergic and GABAergic systems, thereby contributing to neuromodulation of odor information processing.
Collapse
Affiliation(s)
- Emi Kiyokage
- Department of Medical Technology, Kawasaki University of Medical Welfare, Kurashiki, Japan
| | - Satoshi Ichikawa
- Research Center for Ultra-High Voltage Electron Microscopy, Osaka University, Ibaraki, Japan
| | - Sawa Horie
- Department of Anatomy, Kawasaki Medical School, Kurashiki, Japan
| | - Shuichi Hayashi
- Department of Anatomy, Kawasaki Medical School, Kurashiki, Japan
| | - Kazunori Toida
- Research Center for Ultra-High Voltage Electron Microscopy, Osaka University, Ibaraki, Japan.,Department of Anatomy, Kawasaki Medical School, Kurashiki, Japan
| |
Collapse
|
12
|
Manion MTC, Glasper ER, Wang KH. A sex difference in mouse dopaminergic projections from the midbrain to basolateral amygdala. Biol Sex Differ 2022; 13:75. [PMID: 36585727 PMCID: PMC9801632 DOI: 10.1186/s13293-022-00486-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Dopaminergic circuits play important roles in the motivational control of behavior and dysfunction in dopaminergic circuits have been implicated in several psychiatric disorders, such as schizophrenia and depression. While these disorders exhibit different incidence rates in men and women, the potential sex differences in the underlying neural circuits are not well-understood. Previous anatomical tracing studies in mammalian species have revealed a prominent circuit projection connecting the dopaminergic midbrain ventral tegmental area (VTA) to the basolateral amygdala (BLA), which is involved in emotional processing and associative learning. However, whether there is any sex difference in this anatomical circuit remains unknown. METHODS To study the potential sex differences in the VTA-to-BLA dopaminergic circuit, we injected two viral vectors encoding fluorescent reporters of axons and synaptic boutons (AAV-FLEX-tdTomato and AAV-FLEX-SynaptophysinGFP, respectively) into the VTA of a mouse transgenic driver line (tyrosine hydroxylase promoter-driven Cre, or TH-Cre), which restricts the reporter expression to dopaminergic neurons. We then used confocal fluorescent microscopy to image the distribution and density of dopaminergic axons and synaptic boutons in serial sections of both male and female mouse brain. RESULTS We found that the overall labeling intensity of VTA-to-BLA dopaminergic projections is intermediate among forebrain dopaminergic pathways, significantly higher than the projections to the prefrontal cortex, but lower than the projections to the nucleus accumbens. Within the amygdala areas, dopaminergic axons are concentrated in BLA. Although the size of BLA and the density of dopaminergic axons within BLA are similar between male and female mice, the density of dopaminergic synaptic boutons in BLA is significantly higher in male brain than female brain. CONCLUSIONS Our results demonstrate an anatomical sex difference in mouse dopaminergic innervations from the VTA to BLA. This finding may provide a structural foundation to study neural circuit mechanisms underlying sex differences in motivational and emotional behaviors and related psychiatric dysfunctions.
Collapse
Affiliation(s)
- Matthew T. C. Manion
- grid.416868.50000 0004 0464 0574Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892 USA ,grid.164295.d0000 0001 0941 7177Department of Psychology, University of Maryland, College Park, MD 20742 USA ,grid.164295.d0000 0001 0941 7177Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD 20742 USA
| | - Erica R. Glasper
- grid.164295.d0000 0001 0941 7177Department of Psychology, University of Maryland, College Park, MD 20742 USA ,grid.164295.d0000 0001 0941 7177Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD 20742 USA ,grid.261331.40000 0001 2285 7943Department of Neuroscience and Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH 43235 USA
| | - Kuan Hong Wang
- grid.416868.50000 0004 0464 0574Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892 USA ,grid.412750.50000 0004 1936 9166Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY 14642 USA
| |
Collapse
|
13
|
Altered visual cortex excitability in premenstrual dysphoric disorder: Evidence from magnetoencephalographic gamma oscillations and perceptual suppression. PLoS One 2022; 17:e0279868. [PMID: 36584199 PMCID: PMC9803314 DOI: 10.1371/journal.pone.0279868] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/15/2022] [Indexed: 12/31/2022] Open
Abstract
Premenstrual dysphoric disorder (PMDD) is a psychiatric condition characterized by extreme mood shifts during the luteal phase of the menstrual cycle (MC) due to abnormal sensitivity to neurosteroids and unbalanced neural excitation/inhibition (E/I) ratio. We hypothesized that in women with PMDD in the luteal phase, these factors would alter the frequency of magnetoencephalographic visual gamma oscillations, affect modulation of their power by excitatory drive, and decrease perceptual spatial suppression. Women with PMDD and control women were examined twice-during the follicular and luteal phases of their MC. We recorded visual gamma response (GR) while modulating the excitatory drive by increasing the drift rate of the high-contrast grating (static, 'slow', 'medium', and 'fast'). Contrary to our expectations, GR frequency was not affected in women with PMDD in either phase of the MC. GR power suppression, which is normally associated with a switch from the 'optimal' for GR slow drift rate to the medium drift rate, was reduced in women with PMDD and was the only GR parameter that distinguished them from control participants specifically in the luteal phase and predicted severity of their premenstrual symptoms. Over and above the atypical luteal GR suppression, in both phases of the MC women with PMDD had abnormally strong GR facilitation caused by a switch from the 'suboptimal' static to the 'optimal' slow drift rate. Perceptual spatial suppression did not differ between the groups but decreased from the follicular to the luteal phase only in PMDD women. The atypical modulation of GR power suggests that neuronal excitability in the visual cortex is constitutively elevated in PMDD and that this E/I imbalance is further exacerbated during the luteal phase. However, the unaltered GR frequency does not support the hypothesis of inhibitory neuron dysfunction in PMDD.
Collapse
|
14
|
Royo M, Escolano BA, Madrigal MP, Jurado S. AMPA Receptor Function in Hypothalamic Synapses. Front Synaptic Neurosci 2022; 14:833449. [PMID: 35173598 PMCID: PMC8842481 DOI: 10.3389/fnsyn.2022.833449] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 01/03/2022] [Indexed: 12/15/2022] Open
Abstract
AMPA receptors (AMPARs) are critical for mediating glutamatergic synaptic transmission and plasticity, thus playing a major role in the molecular machinery underlying cellular substrates of memory and learning. Their expression pattern, transport and regulatory mechanisms have been extensively studied in the hippocampus, but their functional properties in other brain regions remain poorly understood. Interestingly, electrophysiological and molecular evidence has confirmed a prominent role of AMPARs in the regulation of hypothalamic function. This review summarizes the existing evidence on AMPAR-mediated transmission in the hypothalamus, where they are believed to orchestrate the role of glutamatergic transmission in autonomous, neuroendocrine function, body homeostasis, and social behavior.
Collapse
|
15
|
Bhattacharya S, Amodei R, Vilain E, Roselli CE. Identification of differential hypothalamic DNA methylation and gene expression associated with sexual partner preferences in rams. PLoS One 2022; 17:e0263319. [PMID: 35552544 PMCID: PMC9098078 DOI: 10.1371/journal.pone.0263319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/13/2022] [Indexed: 11/19/2022] Open
Abstract
The sheep is a valuable model to test whether hormone mechanisms that sexually differentiate the brain underlie the expression of sexual partner preferences because as many as 8% of rams prefer same-sex partners. Epigenetic factors such as DNA methylation act as mediators in the interaction between steroid hormones and the genome. Variations in the epigenome could be important in determining morphological or behavior differences among individuals of the same species. In this study, we explored DNA methylation differences in the hypothalamus of male oriented rams (MORs) and female oriented rams (FORs). We employed reduced representation bisulfite sequencing (RRBS) to generate a genome-wide map of DNA methylation and RNA-Seq to profile the transcriptome. We found substantial DNA methylation and gene expression differences between FORs and MORs. Although none of the differentially methylated genes yielded significant functional terms directly associated with sex development, three differentially expressed genes were identified that have been associated previously with sexual behaviors. We hypothesize that these differences are involved in the phenotypic variation in ram sexual partner preferences, whereas future studies will have to find the specific mechanisms. Our results add an intriguing new dimension to sheep behavior that should be useful for further understanding epigenetic and transcriptomic involvement.
Collapse
Affiliation(s)
- Surajit Bhattacharya
- Center for Genetic Medicine Research, Children’s Research Institute, Children’s National Hospital, Washington, DC, United States of America
| | - Rebecka Amodei
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University Portland, Portland, Oregon, United States of America
| | - Eric Vilain
- Center for Genetic Medicine Research, Children’s Research Institute, Children’s National Hospital, Washington, DC, United States of America
| | - Charles E. Roselli
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University Portland, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
16
|
Brann DW, Lu Y, Wang J, Zhang Q, Thakkar R, Sareddy GR, Pratap UP, Tekmal RR, Vadlamudi RK. Brain-derived estrogen and neural function. Neurosci Biobehav Rev 2021; 132:793-817. [PMID: 34823913 PMCID: PMC8816863 DOI: 10.1016/j.neubiorev.2021.11.014] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/26/2021] [Accepted: 11/12/2021] [Indexed: 01/02/2023]
Abstract
Although classically known as an endocrine signal produced by the ovary, 17β-estradiol (E2) is also a neurosteroid produced in neurons and astrocytes in the brain of many different species. In this review, we provide a comprehensive overview of the localization, regulation, sex differences, and physiological/pathological roles of brain-derived E2 (BDE2). Much of what we know regarding the functional roles of BDE2 has come from studies using specific inhibitors of the E2 synthesis enzyme, aromatase, as well as the recent development of conditional forebrain neuron-specific and astrocyte-specific aromatase knockout mouse models. The evidence from these studies support a critical role for neuron-derived E2 (NDE2) in the regulation of synaptic plasticity, memory, socio-sexual behavior, sexual differentiation, reproduction, injury-induced reactive gliosis, and neuroprotection. Furthermore, we review evidence that astrocyte-derived E2 (ADE2) is induced following brain injury/ischemia, and plays a key role in reactive gliosis, neuroprotection, and cognitive preservation. Finally, we conclude by discussing the key controversies and challenges in this area, as well as potential future directions for the field.
Collapse
Affiliation(s)
- Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| | - Yujiao Lu
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Jing Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Roshni Thakkar
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Gangadhara R Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA
| | - Uday P Pratap
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA
| | - Rajeshwar R Tekmal
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA; Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA.
| |
Collapse
|
17
|
Phumsatitpong C, Wagenmaker ER, Moenter SM. Neuroendocrine interactions of the stress and reproductive axes. Front Neuroendocrinol 2021; 63:100928. [PMID: 34171353 PMCID: PMC8605987 DOI: 10.1016/j.yfrne.2021.100928] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 06/17/2021] [Accepted: 06/19/2021] [Indexed: 01/27/2023]
Abstract
Reproduction is controlled by a sequential regulation of the hypothalamo-pituitary-gonadal (HPG) axis. The HPG axis integrates multiple inputs to maintain proper reproductive functions. It has long been demonstrated that stress alters fertility. Nonetheless, the central mechanisms of how stress interacts with the reproductive system are not fully understood. One of the major pathways that is activated during the stress response is the hypothalamo-pituitary-adrenal (HPA) axis. In this review, we discuss several aspects of the interactions between these two neuroendocrine systems to offer insights to mechanisms of how the HPA and HPG axes interact. We have also included discussions of other systems, for example GABA-producing neurons, where they are informative to the overall picture of stress effects on reproduction.
Collapse
Affiliation(s)
- Chayarndorn Phumsatitpong
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Elizabeth R Wagenmaker
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Suzanne M Moenter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
18
|
Knickmeyer RC, Nguyen CT, Young JT, Haunton A, Kosorok MR, Gilmore JH, Styner M, Rothmond DA, Noble PL, Lenroot R, Weickert CS. Impact of gonadectomy on maturational changes in brain volume in adolescent macaques. Psychoneuroendocrinology 2021; 124:105068. [PMID: 33260081 PMCID: PMC8121100 DOI: 10.1016/j.psyneuen.2020.105068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/29/2020] [Accepted: 11/12/2020] [Indexed: 10/23/2022]
Abstract
Adolescence is a transitional period between childhood and adulthood characterized by significant changes in global and regional brain tissue volumes. It is also a period of increasing vulnerability to psychiatric illness. The relationship between these patterns and increased levels of circulating sex steroids during adolescence remains unclear. The objective of the current study was to determine whether gonadectomy, prior to puberty, alters adolescent brain development in male rhesus macaques. Ninety-six structural MRI scans were acquired from 12 male rhesus macaques (8 time points per animal over a two-year period). Six animals underwent gonadectomy and 6 animals underwent a sham operation at 29 months of age. Mixed-effects models were used to determine whether gonadectomy altered developmental trajectories of global and regional brain tissue volumes. We observed a significant effect of gonadectomy on the developmental trajectory of prefrontal gray matter (GM), with intact males showing peak volumes around 3.5 years of age with a subsequent decline. In contrast, prefrontal GM volumes continued to increase in gonadectomized males until the end of the study. We did not observe a significant effect of gonadectomy on prefrontal white matter or on any other global or regional brain tissue volumes, though we cannot rule out that effects might be detected in a larger sample. Results suggest that the prefrontal cortex is more vulnerable to gonadectomy than other brain regions.
Collapse
Affiliation(s)
- Rebecca C. Knickmeyer
- Michigan State University, Institute for Quantitative Health Science and Engineering, Room 2114, Bio Engineering Facility, 775 Woodlot Dr., East Lansing, MI, 48824 USA,University of North Carolina at Chapel Hill, Department of Psychiatry, Campus Box #7160, Chapel Hill, NC 27599-7160, USA
| | - Crystal T. Nguyen
- University of North Carolina at Chapel Hill, Department of Biostatistics, Campus Box #7420, Chapel Hill, NC 27599-7420, USA
| | - Jeffrey T. Young
- University of North Carolina at Chapel Hill, Department of Psychiatry, Campus Box #7160, Chapel Hill, NC 27599-7160, USA
| | - Anne Haunton
- North Carolina School of Science and Mathematics, 1219 Broad St, Durham, NC 27705, USA.
| | - Michael R. Kosorok
- University of North Carolina at Chapel Hill, Department of Biostatistics, Campus Box #7420, Chapel Hill, NC 27599-7420, USA
| | - John H. Gilmore
- University of North Carolina at Chapel Hill, Department of Psychiatry, Campus Box #7160, Chapel Hill, NC 27599-7160, USA
| | - Martin Styner
- University of North Carolina at Chapel Hill, Department of Psychiatry, Campus Box #7160, Chapel Hill, NC 27599-7160, USA; University of North Carolina at Chapel Hill, Department of Computer Science, Campus Box #3175, Chapel Hill, NC 27599-3175, USA.
| | - Debora A. Rothmond
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick NSW 2031, Australia
| | - Pamela L. Noble
- Laboratory of Neuropsychology, National Institute for Mental Health, National Institutes of Health, Bethesda, MD 20892-9663
| | - Rhoshel Lenroot
- University of New Mexico, Department of Psychiatry and Behavioral Sciences, Albuquerque, NM 87131, USA.
| | - Cynthia Shannon Weickert
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia; Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY 13210, USA.
| |
Collapse
|
19
|
Phumsatitpong C, De Guzman RM, Zuloaga DG, Moenter SM. A CRH Receptor Type 1 Agonist Increases GABA Transmission to GnRH Neurons in a Circulating-Estradiol-Dependent Manner. Endocrinology 2020; 161:5892962. [PMID: 32798220 PMCID: PMC7547842 DOI: 10.1210/endocr/bqaa140] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/06/2020] [Indexed: 12/15/2022]
Abstract
GnRH neurons are central regulators of reproduction and respond to factors affecting fertility, such as stress. Corticotropin-releasing hormone (CRH) is released during stress response. In brain slices from unstressed controls, CRH has opposite, estradiol-dependent effects on GnRH neuron firing depending on the CRH receptor activated; activating CRHR-1 stimulates whereas activating CRHR-2 suppresses activity. We investigated possible direct and indirect mechanisms. Mice were ovariectomized and either not treated further (OVX) or given a capsule producing high positive feedback (OVX + E) or low negative feedback (OVX + low E) physiologic circulating estradiol levels. We tested possible direct effects on GnRH neurons by altering voltage-gated potassium currents. Two types of voltage-gated potassium currents (transient IA and sustained IK) were measured; neither CRHR-1 nor CRHR-2 agonists altered potassium current density in GnRH neurons from OVX + E mice. Further, neither CRH nor receptor-specific agonists altered action potential generation in response to current injection in GnRH neurons from OVX + E mice. To test the possible indirect actions, GABAergic postsynaptic currents were monitored. A CRHR-1 agonist increased GABAergic transmission frequency to GnRH neurons from OVX + E, but not OVX, mice, whereas a CRHR-2 agonist had no effect. Finally, we tested if CRH alters the firing rate of arcuate kisspeptin neurons, which provide an important excitatory neuromodulatory input to GnRH neurons. CRH did not acutely alter firing activity of these neurons from OVX, OVX + E or OVX + low E mice. These results suggest CRH increases GnRH neuron activity in an estradiol-dependent manner in part by activating GABAergic afferents. Mechanisms underlying inhibitory effects of CRH remain unknown.
Collapse
Affiliation(s)
| | | | | | - Suzanne M Moenter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, US
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, US
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, US
- Correspondence: Suzanne M. Moenter; 7725 Med Sci II; 1137 E Catherine St; Ann Arbor, MI 48109-5622. E-mail:
| |
Collapse
|
20
|
Ke H, Wu Y, Wang R, Wu X. Creation of a Prognostic Risk Prediction Model for Lung Adenocarcinoma Based on Gene Expression, Methylation, and Clinical Characteristics. Med Sci Monit 2020; 26:e925833. [PMID: 33021972 PMCID: PMC7549534 DOI: 10.12659/msm.925833] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background This study aimed to identify important marker genes in lung adenocarcinoma (LACC) and establish a prognostic risk model to predict the risk of LACC in patients. Material/Methods Gene expression and methylation profiles for LACC and clinical information about cases were downloaded from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases, respectively. Differentially expressed genes (DEGs) and differentially methylated genes (DMGs) between cancer and control groups were selected through meta-analysis. Pearson coefficient correlation analysis was performed to identify intersections between DEGs and DMGs and a functional analysis was performed on the genes that were correlated. Marker genes and clinical factors significantly related to prognosis were identified using univariate and multivariate Cox regression analyses. Risk prediction models were then created based on the marker genes and clinical factors. Results In total, 1975 DEGs and 2095 DMGs were identified. After comparison, 16 prognosis-related genes (EFNB2, TSPAN7, INPP5A, VAMP2, CALML5, SNAI2, RHOBTB1, CKB, ATF7IP2, RIMS2, RCBTB2, YBX1, RAB27B, NFATC1, TCEAL4, and SLC16A3) were selected from 265 overlapping genes. Four clinical factors (pathologic N [node], pathologic T [tumor], pathologic stage, and new tumor) were associated with prognosis. The prognostic risk prediction models were constructed and validated with other independent datasets. Conclusions An integrated model that combines clinical factors and gene markers is useful for predicting risk of LACC in patients. The 16 genes that were identified, including EFNB2, TSPAN7, INPP5A, VAMP2, and CALML5, may serve as novel biomarkers for diagnosis of LACC and prediction of disease prognosis.
Collapse
Affiliation(s)
- Honggang Ke
- Department of Cardiovascular and Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (mainland)
| | - Yunyu Wu
- Qixiu Campus, Nantong University, Nantong, Jiangsu, China (mainland)
| | - Runjie Wang
- Department of Oncology, Wuxi People's Hospital, Wuxi, Jiangsu, China (mainland)
| | - Xiaohong Wu
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University and Wuxi 4th People's Hospital, Wuxi, Jiangsu, China (mainland)
| |
Collapse
|
21
|
Krishna G, Bromberg C, Connell EC, Mian E, Hu C, Lifshitz J, Adelson PD, Thomas TC. Traumatic Brain Injury-Induced Sex-Dependent Changes in Late-Onset Sensory Hypersensitivity and Glutamate Neurotransmission. Front Neurol 2020; 11:749. [PMID: 32849211 PMCID: PMC7419702 DOI: 10.3389/fneur.2020.00749] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/17/2020] [Indexed: 01/15/2023] Open
Abstract
Women approximate one-third of the annual 2.8 million people in the United States who sustain traumatic brain injury (TBI). Several clinical reports support or refute that menstrual cycle-dependent fluctuations in sex hormones are associated with severity of persisting post-TBI symptoms. Previously, we reported late-onset sensory hypersensitivity to whisker stimulation that corresponded with changes in glutamate neurotransmission at 1-month following diffuse TBI in male rats. Here, we incorporated intact age-matched naturally cycling females into the experimental design while monitoring daily estrous cycle. We hypothesized that sex would not influence late-onset sensory hypersensitivity and associated in vivo amperometric extracellular recordings of glutamate neurotransmission within the behaviorally relevant thalamocortical circuit. At 28 days following midline fluid percussion injury (FPI) or sham surgery, young adult Sprague-Dawley rats were tested for hypersensitivity to whisker stimulation using the whisker nuisance task (WNT). As predicted, both male and female rats showed significantly increased sensory hypersensitivity to whisker stimulation after FPI, with females having an overall decrease in whisker nuisance scores (sex effect), but no injury and sex interaction. In males, FPI increased potassium chloride (KCl)-evoked glutamate overflow in primary somatosensory barrel cortex (S1BF) and ventral posteromedial nucleus of the thalamus (VPM), while in females the FPI effect was discernible only within the VPM. Similar to our previous report, we found the glutamate clearance parameters were not influenced by FPI, while a sex-specific effect was evident with female rats showing a lower uptake rate constant both in S1BF and VPM and longer clearance time (in S1BF) in comparison to male rats. Fluctuations in estrous cycle were evident among brain-injured females with longer diestrus (low circulating hormone) phase of the cycle over 28 days post-TBI. Together, these findings add to growing evidence indicating both similarities and differences between sexes in a chronic response to TBI. A better understanding of the influence of gonadal hormones on behavior, neurotransmission, secondary injury and repair processes after TBI is needed both clinically and translationally, with potential impact on acute treatment, rehabilitation, and symptom management.
Collapse
Affiliation(s)
- Gokul Krishna
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Caitlin Bromberg
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Emily Charlotte Connell
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Erum Mian
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Chengcheng Hu
- Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ, United States
| | - Jonathan Lifshitz
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
- Phoenix VA Health Care System, Phoenix, AZ, United States
| | - P. David Adelson
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, AZ, United States
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States
- Phoenix VA Health Care System, Phoenix, AZ, United States
| |
Collapse
|
22
|
Parker EM, Kindja NL, Cheetham CEJ, Sweet RA. Sex differences in dendritic spine density and morphology in auditory and visual cortices in adolescence and adulthood. Sci Rep 2020; 10:9442. [PMID: 32523006 PMCID: PMC7287134 DOI: 10.1038/s41598-020-65942-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/24/2020] [Indexed: 11/24/2022] Open
Abstract
Dendritic spines are small protrusions on dendrites that endow neurons with the ability to receive and transform synaptic input. Dendritic spine number and morphology are altered as a consequence of synaptic plasticity and circuit refinement during adolescence. Dendritic spine density (DSD) is significantly different based on sex in subcortical brain regions associated with the generation of sex-specific behaviors. It is largely unknown if sex differences in DSD exist in auditory and visual brain regions and if there are sex-specific changes in DSD in these regions that occur during adolescent development. We analyzed dendritic spines in 4-week-old (P28) and 12-week-old (P84) male and female mice and found that DSD is lower in female mice due in part to fewer short stubby, long stubby and short mushroom spines. We found striking layer-specific patterns including a significant age by layer interaction and significantly decreased DSD in layer 4 from P28 to P84. Together these data support the possibility of developmental sex differences in DSD in visual and auditory regions and provide evidence of layer-specific refinement of DSD over adolescent brain development.
Collapse
Affiliation(s)
- Emily M Parker
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, USA
- Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, USA
| | - Nathan L Kindja
- Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, USA
| | - Claire E J Cheetham
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, USA
- Center for the Neural Basis of Cognition, Pittsburgh, USA
| | - Robert A Sweet
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, USA.
- Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, USA.
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
23
|
Stefanaki C, Michos A, Latsios G, Tousoulis D, Peppa M, Zosi P, Boschiero D, Bacopoulou F. Sexual Dimorphism of Heart Rate Variability in Adolescence: A Case-Control Study on Depression, Anxiety, Stress Levels, Body Composition, and Heart Rate Variability in Adolescents with Impaired Fasting Glucose. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:2688. [PMID: 32295195 PMCID: PMC7216092 DOI: 10.3390/ijerph17082688] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/10/2020] [Accepted: 04/11/2020] [Indexed: 12/22/2022]
Abstract
Prediabetes in the form of impaired fasting glucose, impaired glucose tolerance, or both is considered as a preliminary stage for the onset of diabetes and diabetic complications. Hormonal fluctuations in adolescence are accompanied by body composition modifications, which are associated with insulin resistance and subclinical inflammation. Bioimpedance (BIA) accurately evaluates body composition, and heart rate variability (HRV) assesses cardiac autonomic function, which are frequently afflicted by insulin resistance. We aimed at evaluating the effect of glycemic status on mental stress, anxiety, and depression status in adolescents with impaired fasting glucose, body composition, and HRV parameters. This is a case-control study to evaluate the effect of the hyperglycemia on depression, anxiety, and stress levels (DASS21 questionnaire), body composition (BIA-ACC-BIOTEKNA©), and HRV (PPG Stress Flow-BIOTEKNA©), between euglycemic adolescents (euglycemic group) and adolescents with impaired fasting glucose (prediabetic group), aged 12-20 years. No differences were found between the prediabetic (n = 13) and the euglycemic (n = 16) groups in the outcome measures, possibly due to the number of participants. Interestingly, females, irrespective of their glycemic status, exhibited altered sympathovagal function as revealed by impaired HRV. In the euglycemic group, HRV parameters were significantly correlated and in line with the DASS21 scores, but in the prediabetic group, similarities to those of adults were observed. Impaired fasting glucose had no impact on mental health, body composition, or HRV parameters in adolescents. HRV parameters were impaired in females, irrespective of their glycemic status. This finding implies that females seem to be more prone to stress disorders, even from a young age. Future studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Charikleia Stefanaki
- Center for Adolescent Medicine and UNESCO Chair on Adolescent Health Care, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, 115 27 Athens, Greece; (A.M.); (F.B.)
- Department of Pediatrics, General Hospital of Nikaia “Agios Panteleimon”, 184 54 Piraeus, Greece;
| | - Athanasios Michos
- Center for Adolescent Medicine and UNESCO Chair on Adolescent Health Care, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, 115 27 Athens, Greece; (A.M.); (F.B.)
| | - George Latsios
- First Cardiology Department, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, 115 28 Athens, Greece; (G.L.); (D.T.)
| | - Dimitrios Tousoulis
- First Cardiology Department, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, 115 28 Athens, Greece; (G.L.); (D.T.)
| | - Melpomeni Peppa
- Endocrine Unit, Second Department of Internal Medicine Propaedeutic, Research Institute and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, 115 28 Athens, Greece;
| | - Paraskevi Zosi
- Department of Pediatrics, General Hospital of Nikaia “Agios Panteleimon”, 184 54 Piraeus, Greece;
| | | | - Flora Bacopoulou
- Center for Adolescent Medicine and UNESCO Chair on Adolescent Health Care, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, 115 27 Athens, Greece; (A.M.); (F.B.)
| |
Collapse
|
24
|
Khbouz B, de Bournonville C, Court L, Taziaux M, Corona R, Arnal JF, Lenfant F, Cornil CA. Role for the membrane estrogen receptor alpha in the sexual differentiation of the brain. Eur J Neurosci 2019; 52:2627-2645. [PMID: 31833601 DOI: 10.1111/ejn.14646] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 10/30/2019] [Accepted: 12/05/2019] [Indexed: 12/25/2022]
Abstract
Estrogens exert pleiotropic effects on multiple physiological and behavioral responses. Male and female sexual behavior in rodents constitutes some of the best-characterized responses activated by estrogens in adulthood and largely depend on ERα. Evidence exists that nucleus- and membrane-initiated estrogen signaling cooperate to orchestrate the activation of these behaviors both in short- and long-term. However, questions remain regarding the mechanism(s) and receptor(s) involved in the early brain programming during development to organize the circuits underlying sexually differentiated responses. Taking advantage of a mouse model harboring a mutation of the ERα palmitoylation site, which prevents membrane ERα signaling (mERα; ERα-C451A), this study investigated the role of mERα on the expression of male and female sexual behavior and neuronal populations that differ between sexes. The results revealed no genotype effect on the expression of female sexual behavior, while male sexual behavior was significantly reduced, but not abolished, in males homozygous for the mutation. Similarly, the number of kisspeptin- (Kp-ir) and calbindin-immunoreactive (Cb-ir) neurons in the anteroventral periventricular nucleus (AVPv) and the sexually dimorphic nucleus of the preoptic area (SDN-POA), respectively, were not different between genotypes in females. In contrast, homozygous males showed increased numbers of Kp-ir and decreased numbers of Cb-ir neurons compared to wild-types, thus leading to an intermediate phenotype between females and wild-type males. Importantly, females neonatally treated with estrogens exhibited the same neurochemical phenotype as their corresponding genotype among males. Together, these data provide evidence that mERα is involved in the perinatal programming of the male brain.
Collapse
Affiliation(s)
- Badr Khbouz
- GIGA Neurosciences, University of Liège, Liège, Belgium
| | | | - Lucas Court
- GIGA Neurosciences, University of Liège, Liège, Belgium
| | | | - Rebeca Corona
- GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Jean-François Arnal
- INSERM/UPS UMR 1048-I2MC, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Françoise Lenfant
- INSERM/UPS UMR 1048-I2MC, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | | |
Collapse
|
25
|
Wang Y, Wu H, Sun ZS. The biological basis of sexual orientation: How hormonal, genetic, and environmental factors influence to whom we are sexually attracted. Front Neuroendocrinol 2019; 55:100798. [PMID: 31593707 DOI: 10.1016/j.yfrne.2019.100798] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/29/2019] [Accepted: 10/03/2019] [Indexed: 12/11/2022]
Abstract
Humans develop relatively stable attractions to sexual partners during maturation and present a spectrum of sexual orientation from homosexuality to heterosexuality encompassing varying degrees of bisexuality, with some individuals also displaying asexuality. Sexual orientation represents a basic life phenomenon for humans. However, the molecular mechanisms underlying these diverse traits of sexual orientation remain highly controversial. In this review, we systematically discuss recent advancements in sexual orientation research, including those related to measurements and associated brain regions. Current findings regarding sexual orientation modulation by hormonal, genetic, maternal immune system, and environmental factors are summarized in both human and model systems. We also emphasize that future studies should recognize the differences between males and females and pay more attention to minor traits and the epigenetic regulation of sexual orientation. A comprehensive view of sexual orientation may promote our understanding of the biological basis of sex, and that of human reproduction, and evolution.
Collapse
Affiliation(s)
- Yan Wang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Haoda Wu
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; Sino-Danish College, University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Zhong Sheng Sun
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; Sino-Danish College, University of the Chinese Academy of Sciences, Beijing 100190, China; Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
26
|
Grafe LA, Geng E, Corbett B, Urban K, Bhatnagar S. Sex- and Stress-Dependent Effects on Dendritic Morphology and Spine Densities in Putative Orexin Neurons. Neuroscience 2019; 418:266-278. [PMID: 31442567 DOI: 10.1016/j.neuroscience.2019.08.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/11/2019] [Accepted: 08/13/2019] [Indexed: 01/23/2023]
Abstract
We recently found that non-stressed female rats have higher basal prepro-orexin expression and activation of orexinergic neurons compared to non-stressed males, which lead to impaired habituation to repeated restraint stress at the behavioral, neural, and endocrine level. Here, we extended our study of sex differences in the orexin system by examining spine densities and dendritic morphology in putative orexin neurons in adult male and female rats that were exposed to 5 consecutive days of 30-min restraint. Analysis of spine distribution and density indicated that putative orexinergic neurons in control non-stressed females had significantly more dendritic spines than those in control males, and the majority of these were mushroom spines. This morphological finding may suggest more excitatory input onto orexin neurons in female rats. As orexin neurons are known to promote the hypothalamic-pituitary-adrenal response, this morphological change in orexin neurons could underlie the impaired habituation to repeated stress in female rats. Dendritic complexity did not differ between non-stressed males and females, however repeated restraint stress decreased total dendritic length, nodes, and branching primarily in males. Thus, reduced dendritic complexity of putative orexinergic neurons is observed in males but not in females after 5days of repeated restraint stress. This morphological change might be reflective of decreased orexin system function, which may allow males to habituate more fully to repeated restraint than females. These results extend our understanding of the role of orexin neurons in regulating habituation and demonstrate changes in putative orexin cell morphology and spines that may underlie sex differences in habituation.
Collapse
Affiliation(s)
- Laura A Grafe
- Department of Psychology, Bryn Mawr College, Bryn Mawr, PA 19010, USA
| | - Eric Geng
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Brian Corbett
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kimberly Urban
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Seema Bhatnagar
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
27
|
Liang SL. The glutamine-glutamate cycle regulates synaptic glutamate release in the ventrolateral ventromedial nucleus of the hypothalamus of perinatal female rats. J Neuroendocrinol 2018; 30:e12642. [PMID: 30168642 DOI: 10.1111/jne.12642] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/23/2018] [Accepted: 08/27/2018] [Indexed: 12/11/2022]
Abstract
The astrocytic glutamine (Gln)-glutamate (Glu) cycle (GGC) supplies Gln for the regulation of glutamatergic synaptic transmission (GST) in the adult hippocampus. Increased synaptic Glu release in the perinatal ventrolateral ventromedial nucleus of the hypothalamus (vlVMH) modulates sexual differentiation, however, whether GGC regulates GST in the perinatal vlVMH has not been determined. Sex differences in oestradiol (E2 ) levels exist in the neonatal hypothalamus, and E2 increases levels of glutamine synthetase and glutaminase, two key enzymes involved in the GGC. Thus, it is hypothesised that sexually dimorphic phenotypes may exist in glutamatergic synapses associated with the GGC in the vlVMH in perinatal rats. Whole-cell voltage-clamp recordings in vlVMH neurones in brain slices from male and female pups revealed that pharmacological disruption of the GGC by α-(methylamino) isobutyric acid (5 mmol L-1 ), which blocks neuronal Gln uptake; or by l-methionine sulphoximine (1.5 mmol L-1 ), which inhibits astrocytic Gln synthesis, decreased miniature excitatory postsynaptic current (mEPSC) amplitudes in female but not male pups. By contrast, GGC interruptions decreased evoked (e)EPSC amplitudes in both sexes following increased synaptic activity produced by a period of stimulation. In male pups, the decreased eEPSCs were attributable to reduced Glu release, as assessed by paired-pulse stimulations, whereas, in female pups, they were attributable to decreased Glu content in the synaptic vesicles, as measured by strontium-evoked mEPSCs. The l-methionine sulphoximine-mediated decrease in eEPSCs was rapidly rescued by exogenous Gln in female but not male pups. The reductions in mEPSCs and eEPSCs in female pups were accompanied by enhanced blocking effects of the low-affinity Glu AMPA receptor antagonist, γ-d-glutamylglycine, consistent with diminished Glu release. In conclusion, female, but not male pups, rely on constitutive astrocytic Gln for sustained synaptic Glu release in the vlVMH. This glutamatergic synaptic phenotype may be associated with brain and behaviour feminisation and/or defeminisation in rats.
Collapse
Affiliation(s)
- Shu-Ling Liang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Linkou, Tao-Yuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, Tao-Yuan, Taiwan
| |
Collapse
|
28
|
Jardí F, Laurent MR, Dubois V, Kim N, Khalil R, Decallonne B, Vanderschueren D, Claessens F. Androgen and estrogen actions on male physical activity: a story beyond muscle. J Endocrinol 2018; 238:R31-R52. [PMID: 29743340 DOI: 10.1530/joe-18-0125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/09/2018] [Indexed: 12/15/2022]
Abstract
Physical inactivity is a pandemic that contributes to several chronic diseases and poses a significant burden on health care systems worldwide. The search for effective strategies to combat sedentary behavior has led to an intensification of the research efforts to unravel the biological substrate controlling activity. A wide body of preclinical evidence makes a strong case for sex steroids regulating physical activity in both genders, albeit the mechanisms implicated remain unclear. The beneficial effects of androgens on muscle as well as on other peripheral functions might play a role in favoring adaptation to exercise. Alternatively or in addition, sex steroids could act on specific brain circuitries to boost physical activity. This review critically discusses the evidence supporting a role for androgens and estrogens stimulating male physical activity, with special emphasis on the possible role of peripheral and/or central mechanisms. Finally, the potential translation of these findings to humans is briefly discussed.
Collapse
Affiliation(s)
- Ferran Jardí
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Michaël R Laurent
- Molecular Endocrinology LaboratoryDepartment of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Gerontology and GeriatricsDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Vanessa Dubois
- Molecular Endocrinology LaboratoryDepartment of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Nari Kim
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Rougin Khalil
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Brigitte Decallonne
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Frank Claessens
- Molecular Endocrinology LaboratoryDepartment of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
29
|
Wang L, Burger LL, Greenwald-Yarnell ML, Myers MG, Moenter SM. Glutamatergic Transmission to Hypothalamic Kisspeptin Neurons Is Differentially Regulated by Estradiol through Estrogen Receptor α in Adult Female Mice. J Neurosci 2018; 38:1061-1072. [PMID: 29114074 PMCID: PMC5792470 DOI: 10.1523/jneurosci.2428-17.2017] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 09/28/2017] [Accepted: 10/30/2017] [Indexed: 01/20/2023] Open
Abstract
Estradiol feedback regulates gonadotropin-releasing hormone (GnRH) neurons and subsequent luteinizing hormone (LH) release. Estradiol acts via estrogen receptor α (ERα)-expressing afferents of GnRH neurons, including kisspeptin neurons in the anteroventral periventricular (AVPV) and arcuate nuclei, providing homeostatic feedback on episodic GnRH/LH release as well as positive feedback to control ovulation. Ionotropic glutamate receptors are important for estradiol feedback, but it is not known where they fit in the circuitry. Estradiol-negative feedback decreased glutamatergic transmission to AVPV and increased it to arcuate kisspeptin neurons; positive feedback had the opposite effect. Deletion of ERα in kisspeptin cells decreased glutamate transmission to AVPV neurons and markedly increased it to arcuate kisspeptin neurons, which also exhibited increased spontaneous firing rate. KERKO mice had increased LH pulse frequency, indicating loss of negative feedback. These observations indicate that ERα in kisspeptin cells is required for appropriate differential regulation of these neurons and neuroendocrine output by estradiol.SIGNIFICANCE STATEMENT The brain regulates fertility through gonadotropin-releasing hormone (GnRH) neurons. Ovarian estradiol regulates the pattern of GnRH (negative feedback) and initiates a surge of release that triggers ovulation (positive feedback). GnRH neurons do not express the estrogen receptor needed for feedback (estrogen receptor α [ERα]); kisspeptin neurons in the arcuate and anteroventral periventricular nuclei are postulated to mediate negative and positive feedback, respectively. Here we extend the network through which feedback is mediated by demonstrating that glutamatergic transmission to these kisspeptin populations is differentially regulated during the reproductive cycle and by estradiol. Electrophysiological and in vivo hormone profile experiments on kisspeptin-specific ERα knock-out mice demonstrate that ERα in kisspeptin cells is required for appropriate differential regulation of these neurons and for neuroendocrine output.
Collapse
Affiliation(s)
- Luhong Wang
- Departments of Molecular and Integrative Physiology
| | | | | | - Martin G Myers
- Departments of Molecular and Integrative Physiology
- Internal Medicine
- Michigan Diabetes Research & Training Center, University of Michigan, Ann Arbor, Michigan 48109
| | - Suzanne M Moenter
- Departments of Molecular and Integrative Physiology,
- Obstetrics and Gynecology
- Internal Medicine
| |
Collapse
|
30
|
Turano A, Osborne BF, Schwarz JM. Sexual Differentiation and Sex Differences in Neural Development. Curr Top Behav Neurosci 2018; 43:69-110. [PMID: 29967999 DOI: 10.1007/7854_2018_56] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Sex determination occurs at the moment of conception, as a result of XX or XY chromosome pairing. From that point, the body undergoes the process of sexual differentiation, inducing the development of physical characteristics that are easily distinguishable between the sexes and are often reflected in one's physical appearance and gender identity. Although less apparent, the brain also undergoes sexual differentiation. Sex differences in the brain are organized during a critical period of neural development and have an instrumental role in determining the physiology and behavior of an individual throughout the lifespan. Understanding the extent of sex differences in neurodevelopment also influences our understanding of the potential risk for a number of neurodevelopmental, neurological, and mental health disorders that exhibit strong sex biases. Advances made in our understanding of sexually dimorphic brain nuclei, sex differences in neural cell communication, and sex differences in the communication between the brain and peripheral organs are all research fields that have provided valuable information related to the physiological and behavioral outcomes of sex differences in brain development. More recently, investigations into the impact of epigenetic mechanisms on sexual differentiation of the brain have indicated that changes in gene expression, via epigenetic modifications, also contribute to sexual differentiation of the developing brain. Still, there are a number of important questions and ideas that have arisen from our current understanding of sex differences in neurodevelopmental processes that necessitate more time and attention in this field.
Collapse
Affiliation(s)
- Alexandra Turano
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, USA
| | - Brittany F Osborne
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, USA
| | - Jaclyn M Schwarz
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, USA.
| |
Collapse
|
31
|
Phumsatitpong C, Moenter SM. Estradiol-Dependent Stimulation and Suppression of Gonadotropin-Releasing Hormone Neuron Firing Activity by Corticotropin-Releasing Hormone in Female Mice. Endocrinology 2018; 159:414-425. [PMID: 29069304 PMCID: PMC5761586 DOI: 10.1210/en.2017-00747] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/17/2017] [Indexed: 11/19/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons are the final central regulators of reproduction, integrating various inputs that modulate fertility. Stress typically inhibits reproduction but can be stimulatory; stress effects can also be modulated by steroid milieu. Corticotropin-releasing hormone (CRH) released during the stress response may suppress reproduction independent of downstream glucocorticoids. We hypothesized CRH suppresses fertility by decreasing GnRH neuron firing activity. To test this, mice were ovariectomized (OVX) and either implanted with an estradiol capsule (OVX+E) or not treated further to examine the influence of estradiol on GnRH neuron response to CRH. Targeted extracellular recordings were used to record firing activity from green fluorescent protein-identified GnRH neurons in brain slices before and during CRH treatment; recordings were done in the afternoon when estradiol has a positive feedback effect to increase GnRH neuron firing. In OVX mice, CRH did not affect the firing rate of GnRH neurons. In contrast, CRH exhibited dose-dependent stimulatory (30 nM) or inhibitory (100 nM) effects on GnRH neuron firing activity in OVX+E mice; both effects were reversible. The dose-dependent effects of CRH appear to result from activation of different receptor populations; a CRH receptor type-1 agonist increased firing activity in GnRH neurons, whereas a CRH receptor type-2 agonist decreased firing activity. CRH and specific agonists also differentially regulated short-term burst frequency and burst properties, including burst duration, spikes/burst, and/or intraburst interval. These results indicate that CRH alters GnRH neuron activity and that estradiol is required for CRH to exert both stimulatory and inhibitory effects on GnRH neurons.
Collapse
Affiliation(s)
- Chayarndorn Phumsatitpong
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Suzanne M. Moenter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
32
|
Oyola MG, Handa RJ. Hypothalamic-pituitary-adrenal and hypothalamic-pituitary-gonadal axes: sex differences in regulation of stress responsivity. Stress 2017; 20:476-494. [PMID: 28859530 PMCID: PMC5815295 DOI: 10.1080/10253890.2017.1369523] [Citation(s) in RCA: 425] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gonadal hormones play a key role in the establishment, activation, and regulation of the hypothalamic-pituitary-adrenal (HPA) axis. By influencing the response and sensitivity to releasing factors, neurotransmitters, and hormones, gonadal steroids help orchestrate the gain of the HPA axis to fine-tune the levels of stress hormones in the general circulation. From early life to adulthood, gonadal steroids can differentially affect the HPA axis, resulting in sex differences in the responsivity of this axis. The HPA axis influences many physiological functions making an organism's response to changes in the environment appropriate for its reproductive status. Although the acute HPA response to stressors is a beneficial response, constant activation of this circuitry by chronic or traumatic stressful episodes may lead to a dysregulation of the HPA axis and cause pathology. Compared to males, female mice and rats show a more robust HPA axis response, as a result of circulating estradiol levels which elevate stress hormone levels during non-threatening situations, and during and after stressors. Fluctuating levels of gonadal steroids in females across the estrous cycle are a major factor contributing to sex differences in the robustness of HPA activity in females compared to males. Moreover, gonadal steroids may also contribute to epigenetic and organizational influences on the HPA axis even before puberty. Correspondingly, crosstalk between the hypothalamic-pituitary-gonadal (HPG) and HPA axes could lead to abnormalities of stress responses. In humans, a dysregulated stress response is one of the most common symptoms seen across many neuropsychiatric disorders, and as a result, such interactions may exacerbate peripheral pathologies. In this review, we discuss the HPA and HPG axes and review how gonadal steroids interact with the HPA axis to regulate the stress circuitry during all stages in life.
Collapse
Affiliation(s)
- Mario G Oyola
- a Department of Biomedical Sciences , Colorado State University , Fort Collins , CO , USA
| | - Robert J Handa
- a Department of Biomedical Sciences , Colorado State University , Fort Collins , CO , USA
| |
Collapse
|
33
|
Neuroimmunology and neuroepigenetics in the establishment of sex differences in the brain. Nat Rev Neurosci 2017. [PMID: 28638119 DOI: 10.1038/nrn.2017.61] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The study of sex differences in the brain is a topic of neuroscientific study that has broad reaching implications for culture, society and biomedical science. Recent research in rodent models has led to dramatic shifts in our views of the mechanisms underlying the sexual differentiation of the brain. These include the surprising discoveries of a role for immune cells and inflammatory mediators in brain masculinization and a role for epigenetic suppression in brain feminization. How and to what degree these findings will translate to human brain development will be questions of central importance in future research in this field.
Collapse
|
34
|
Nesan D, Kurrasch DM. Genetic programs of the developing tuberal hypothalamus and potential mechanisms of their disruption by environmental factors. Mol Cell Endocrinol 2016; 438:3-17. [PMID: 27720896 DOI: 10.1016/j.mce.2016.09.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/22/2016] [Accepted: 09/29/2016] [Indexed: 12/15/2022]
Abstract
The hypothalamus is a critical regulator of body homeostasis, influencing the autonomic nervous system and releasing trophic hormones to modulate the endocrine system. The developmental mechanisms that govern formation of the mature hypothalamus are becoming increasingly understood as research in this area grows, leading us to gain appreciation for how these developmental programs are susceptible to disruption by maternal exposure to endocrine disrupting chemicals or other environmental factors in utero. These vulnerabilities, combined with the prominent roles of the various hypothalamic nuclei in regulating appetite, reproductive behaviour, mood, and other physiologies, create a window whereby early developmental disruption can have potent long-term effects. Here we broadly outline our current understanding of hypothalamic development, with a particular focus on the tuberal hypothalamus, including what is know about nuclear coalescing and maturation. We finish by discussing how exposure to environmental or maternally-derived factors can perhaps disrupt these hypothalamic developmental programs, and potentially lead to neuroendocrine disease states.
Collapse
Affiliation(s)
- Dinushan Nesan
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
35
|
McInnis CM, Venu S, Park JH. Steroid-independent male sexual behavior in B6D2F2 male mice. Horm Behav 2016; 85:26-29. [PMID: 27476435 DOI: 10.1016/j.yhbeh.2016.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/19/2016] [Accepted: 07/27/2016] [Indexed: 11/16/2022]
Abstract
It is well established that male sexual behavior (MSB) is regulated by gonadal steroids; however, individual differences in MSB, independent of gonadal steroids, are prevalent across a wide range of species, and further investigation is necessary to advance our understanding of steroid-independent MSB. Studies utilizing B6D2F1 hybrid male mice in which a significant proportion retain MSB after long-term orchidectomy, identified as steroid-independent-maters (SI-maters), have begun to unravel the genetic underpinnings of steroid-independent MSB. A recent study demonstrated that steroid-independent MSB is a heritable behavioral phenotype that is mainly passed down from B6D2F1 hybrid SI-maters when crossed with C57BL6J female mice. To begin to uncover whether the strain of the dam plays a role in the inheritance of steroid-independent MSB, B6D2F1 hybrid females were crossed with B6D2F1 hybrid males. While the present study confirms the finding that steroid-independent MSB is a heritable behavioral phenotype and that SI-mater sires are more likely to pass down some components of MSB than SI-non-maters to their offspring, it also reveals that the B6D2F2 male offspring that were identified as SI-maters that displayed the full repertoire of steroid-independent MSB had the same probability of being sired from either a B6D2F1 SI-mater or SI-non-mater. These results, in conjunction with previous findings, indicate that the specific chromosomal loci pattern that codes for steroid-independent MSB in the B6D2F2 male offspring may result regardless of whether the father was a SI-mater or SI-non-mater, and that the maternal strain may be an important factor in the inheritance of steroid-independent MSB.
Collapse
Affiliation(s)
- Christine M McInnis
- Psychology Department, University of Massachusetts, Boston, Boston, MA 02125, United States.
| | - Samitha Venu
- Psychology Department, University of Massachusetts, Boston, Boston, MA 02125, United States
| | - Jin Ho Park
- Psychology Department, University of Massachusetts, Boston, Boston, MA 02125, United States
| |
Collapse
|
36
|
Gonadal hormone modulation of intracellular calcium as a mechanism of neuroprotection. Front Neuroendocrinol 2016; 42:40-52. [PMID: 26930421 DOI: 10.1016/j.yfrne.2016.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 02/22/2016] [Accepted: 02/26/2016] [Indexed: 12/28/2022]
Abstract
Hormones have wide-ranging effects throughout the nervous system, including the ability interact with and modulate many aspects of intracellular calcium regulation and calcium signaling. Indeed, these interactions specifically may help to explain the often opposing or paradoxical effects of hormones, such as their ability to both promote and prevent neuronal cell death during development, as well as reduce or exacerbate damage following an insult or injury in adulthood. Here, we review the basic mechanisms underlying intracellular calcium regulation-perhaps the most dynamic and flexible of all signaling molecules-and discuss how gonadal hormones might manipulate these mechanisms to coordinate diverse cellular responses and achieve disparate outcomes. Additional future research that specifically addresses questions of sex and hormone effects on calcium signaling at different ages will be critical to understanding hormone-mediated neuroprotection.
Collapse
|
37
|
McInnis CM, Bonthuis PJ, Rissman EF, Park JH. Inheritance of steroid-independent male sexual behavior in male offspring of B6D2F1 mice. Horm Behav 2016; 80:132-138. [PMID: 26940434 PMCID: PMC4818728 DOI: 10.1016/j.yhbeh.2016.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 02/23/2016] [Accepted: 02/24/2016] [Indexed: 01/28/2023]
Abstract
The importance of gonadal steroids in modulating male sexual behavior is well established. Individual differences in male sexual behavior, independent of gonadal steroids, are prevalent across a wide range of species, including man. However, the genetic mechanisms underlying steroid-independent male sexual behavior are poorly understood. A high proportion of B6D2F1 hybrid male mice demonstrates steroid-independent male sexual behavior (identified as "maters"), providing a mouse model that opens up avenues of investigation into the mechanisms regulating male sexual behavior in the absence of gonadal hormones. Recent studies have revealed several proteins that play a significant factor in regulating steroid-independent male sexual behavior in B6D2F1 male mice, including amyloid precursor protein (APP), tau, and synaptophysin. The specific goals of our study were to determine whether steroid-independent male sexual behavior was a heritable trait by determining if it was dependent upon the behavioral phenotype of the B6D2F1 sire, and whether the differential expression of APP, tau, and synaptophysin in the medial preoptic area found in the B6D2F1 sires that did and did not mate after gonadectomy was similar to those found in their male offspring. After adult B6D2F1 male mice were bred with C57BL/6J female mice, they and their male offspring (BXB1) were orchidectomized and identified as either maters or "non-maters". A significant proportion of the BXB1 maters was sired only from B6D2F1 maters, indicating that the steroid-independent male sexual behavior behavioral phenotype of the B6D2F1 hybrid males, when crossed with C57BL/6J female mice, is inherited by their male offspring. Additionally, APP, tau, and synaptophysin were elevated in in the medial preoptic area in both the B6D2F1 and BXB1 maters relative to the B6D2F1 and BXB1 non-maters, respectively, suggesting a potential genetic mechanism for the inheritance of steroid-independent male sexual behavior.
Collapse
Affiliation(s)
- Christine M McInnis
- Psychology Department, University of Massachusetts, Boston, Boston, MA 02125, United States.
| | - Paul J Bonthuis
- Department of Biochemistry & Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, United States
| | - Emilie F Rissman
- Department of Biochemistry & Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, United States
| | - Jin Ho Park
- Psychology Department, University of Massachusetts, Boston, Boston, MA 02125, United States; Department of Biochemistry & Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, United States
| |
Collapse
|
38
|
Lim WL, Idris MM, Kevin FS, Soga T, Parhar IS. Maternal Dexamethasone Exposure Alters Synaptic Inputs to Gonadotropin-Releasing Hormone Neurons in the Early Postnatal Rat. Front Endocrinol (Lausanne) 2016; 7:117. [PMID: 27630615 PMCID: PMC5005956 DOI: 10.3389/fendo.2016.00117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 08/11/2016] [Indexed: 11/18/2022] Open
Abstract
Maternal dexamethasone [(DEX); a glucocorticoid receptor agonist] exposure delays pubertal onset and alters reproductive behavior in the adult offspring. However, little is known whether maternal DEX exposure affects the offspring's reproductive function by disrupting the gonadotropin-releasing hormone (GnRH) neuronal function in the brain. Therefore, this study determined the exposure of maternal DEX on the GnRH neuronal spine development and synaptic cluster inputs to GnRH neurons using transgenic rats expressing enhanced green fluorescent protein (EGFP) under the control of GnRH promoter. Pregnant females were administered with DEX (0.1 mg/kg) or vehicle (VEH, water) daily during gestation day 13-20. Confocal imaging was used to examine the spine density of EGFP-GnRH neurons by three-dimensional rendering and synaptic cluster inputs to EGFP-GnRH neurons by synapsin I immunohistochemistry on postnatal day 0 (P0) males. The spine morphology and number on GnRH neurons did not change between the P0 males following maternal DEX and VEH treatment. The number of synaptic clusters within the organum vasculosum of the lamina terminalis (OVLT) was decreased by maternal DEX exposure in P0 males. Furthermore, the number and levels of synaptic cluster inputs in close apposition with GnRH neurons was decreased following maternal DEX exposure in the OVLT region of P0 males. In addition, the postsynaptic marker molecule, postsynaptic density 95, was observed in GnRH neurons following both DEX and VEH treatment. These results suggest that maternal DEX exposure alters neural afferent inputs to GnRH neurons during early postnatal stage, which could lead to reproductive dysfunction during adulthood.
Collapse
Affiliation(s)
- Wei Ling Lim
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya, Malaysia
| | - Marshita Mohd Idris
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya, Malaysia
| | - Felix Suresh Kevin
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya, Malaysia
| | - Tomoko Soga
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya, Malaysia
- *Correspondence: Tomoko Soga,
| | - Ishwar S. Parhar
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya, Malaysia
| |
Collapse
|
39
|
Forger NG, Strahan JA, Castillo-Ruiz A. Cellular and molecular mechanisms of sexual differentiation in the mammalian nervous system. Front Neuroendocrinol 2016; 40:67-86. [PMID: 26790970 PMCID: PMC4897775 DOI: 10.1016/j.yfrne.2016.01.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 12/31/2015] [Accepted: 01/09/2016] [Indexed: 01/16/2023]
Abstract
Neuroscientists are likely to discover new sex differences in the coming years, spurred by the National Institutes of Health initiative to include both sexes in preclinical studies. This review summarizes the current state of knowledge of the cellular and molecular mechanisms underlying sex differences in the mammalian nervous system, based primarily on work in rodents. Cellular mechanisms examined include neurogenesis, migration, the differentiation of neurochemical and morphological cell phenotype, and cell death. At the molecular level we discuss evolving roles for epigenetics, sex chromosome complement, the immune system, and newly identified cell signaling pathways. We review recent findings on the role of the environment, as well as genome-wide studies with some surprising results, causing us to re-think often-used models of sexual differentiation. We end by pointing to future directions, including an increased awareness of the important contributions of tissues outside of the nervous system to sexual differentiation of the brain.
Collapse
Affiliation(s)
- Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States.
| | - J Alex Strahan
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States.
| | | |
Collapse
|
40
|
McCarthy MM, Pickett LA, VanRyzin JW, Kight KE. Surprising origins of sex differences in the brain. Horm Behav 2015; 76:3-10. [PMID: 25917865 PMCID: PMC4620061 DOI: 10.1016/j.yhbeh.2015.04.013] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 03/22/2015] [Accepted: 04/06/2015] [Indexed: 11/22/2022]
Abstract
This article is part of a Special Issue "SBN 2014". Discerning the biologic origins of neuroanatomical sex differences has been of interest since they were first reported in the late 60's and early 70's. The centrality of gonadal hormone exposure during a developmental critical window cannot be denied but hormones are indirect agents of change, acting to induce gene transcription or modulate membrane bound signaling cascades. Sex differences in the brain include regional volume differences due to differential cell death, neuronal and glial genesis, dendritic branching and synaptic patterning. Early emphasis on mechanism therefore focused on neurotransmitters and neural growth factors, but by and large these endpoints failed to explain the origins of neural sex differences. More recently evidence has accumulated in favor of inflammatory mediators and immune cells as principle regulators of brain sexual differentiation and reveal that the establishment of dimorphic circuits is not cell autonomous but instead requires extensive cell-to-cell communication including cells of non-neuronal origin. Despite the multiplicity of cells involved the nature of the sex differences in the neuroanatomical endpoints suggests canalization, a process that explains the robustness of individuals in the face of intrinsic and extrinsic variability. We propose that some neuroanatomical endpoints are canalized to enhance sex differences in the brain by reducing variability within one sex while also preventing the sexes from diverging too greatly. We further propose mechanisms by which such canalization could occur and discuss what relevance this may have to sex differences in behavior.
Collapse
Affiliation(s)
- Margaret M McCarthy
- Department of Pharmacology, Program in Neuroscience and Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Lindsay A Pickett
- Department of Pharmacology, Program in Neuroscience and Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jonathan W VanRyzin
- Department of Pharmacology, Program in Neuroscience and Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Katherine E Kight
- Department of Pharmacology, Program in Neuroscience and Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
41
|
Rainville J, Pollard K, Vasudevan N. Membrane-initiated non-genomic signaling by estrogens in the hypothalamus: cross-talk with glucocorticoids with implications for behavior. Front Endocrinol (Lausanne) 2015; 6:18. [PMID: 25762980 PMCID: PMC4329805 DOI: 10.3389/fendo.2015.00018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 01/30/2015] [Indexed: 12/12/2022] Open
Abstract
The estrogen receptor and glucocorticoid receptor are members of the nuclear receptor superfamily that can signal using both non-genomic and genomic transcriptional modes. Though genomic modes of signaling have been well characterized and several behaviors attributed to this signaling mechanism, the physiological significance of non-genomic modes of signaling has not been well understood. This has partly been due to the controversy regarding the identity of the membrane ER (mER) or membrane GR (mGR) that may mediate rapid, non-genomic signaling and the downstream signaling cascades that may result as a consequence of steroid ligands binding the mER or the mGR. Both estrogens and glucocorticoids exert a number of actions on the hypothalamus, including feedback. This review focuses on the various candidates for the mER or mGR in the hypothalamus and the contribution of non-genomic signaling to classical hypothalamically driven behaviors and changes in neuronal morphology. It also attempts to categorize some of the possible functions of non-genomic signaling at both the cellular level and at the organismal level that are relevant for behavior, including some behaviors that are regulated by both estrogens and glucocorticoids in a potentially synergistic manner. Lastly, it attempts to show that steroid signaling via non-genomic modes may provide the organism with rapid behavioral responses to stimuli.
Collapse
Affiliation(s)
- Jennifer Rainville
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, USA
| | - Kevin Pollard
- Neuroscience Program, Tulane University, New Orleans, LA, USA
| | - Nandini Vasudevan
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, USA
- Neuroscience Program, Tulane University, New Orleans, LA, USA
- *Correspondence: Nandini Vasudevan, Department of Cell and Molecular Biology, Tulane University, 2000 Percival Stern Hall, New Orleans, LA 70118, USA e-mail:
| |
Collapse
|
42
|
Kight KE, McCarthy MM. Using sex differences in the developing brain to identify nodes of influence for seizure susceptibility and epileptogenesis. Neurobiol Dis 2014; 72 Pt B:136-43. [PMID: 24892888 PMCID: PMC5322568 DOI: 10.1016/j.nbd.2014.05.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/12/2014] [Accepted: 05/22/2014] [Indexed: 12/12/2022] Open
Abstract
Sexual differentiation of the developing brain organizes the neural architecture differently between males and females, and the main influence on this process is exposure to gonadal steroids during sensitive periods of prenatal and early postnatal development. Many molecular and cellular processes are influenced by steroid hormones in the developing brain, including gene expression, cell birth and death, neurite outgrowth and synaptogenesis, and synaptic activity. Perturbations in these processes can alter neuronal excitability and circuit activity, leading to increased seizure susceptibility and the promotion of pathological processes that constitute epileptogenesis. In this review, we will provide a general overview of sex differences in the early developing brain that may be relevant for altered seizure susceptibility in early life, focusing on limbic areas of the brain. Sex differences that have the potential to alter the progress of epileptogenesis are evident at molecular and cellular levels in the developing brain, and include differences in neuronal excitability, response to environmental insult, and epigenetic control of gene expression. Knowing how these processes differ between the sexes can help us understand fundamental mechanisms underlying gender differences in seizure susceptibility and epileptogenesis.
Collapse
Affiliation(s)
- Katherine E Kight
- Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Margaret M McCarthy
- Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Pharmacology and Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
43
|
Olivetti PR, Maheshwari A, Noebels JL. Neonatal estradiol stimulation prevents epilepsy in Arx model of X-linked infantile spasms syndrome. Sci Transl Med 2014; 6:220ra12. [PMID: 24452264 DOI: 10.1126/scitranslmed.3007231] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Infantile spasms are a catastrophic form of pediatric epilepsy with inadequate treatment. In patients, mutation of ARX, a transcription factor selectively expressed in neuronal precursors and adult inhibitory interneurons, impairs cell migration and causes a major inherited subtype of the disease X-linked infantile spasms syndrome. Using an animal model, the Arx((GCG)10+7) mouse, we determined that brief estradiol (E2) administration during early postnatal development prevented spasms in infancy and seizures in adult mutants. E2 was ineffective when delivered after puberty or 30 days after birth. Early E2 treatment altered mRNA levels of three downstream targets of Arx (Shox2, Ebf3, and Lgi1) and restored depleted interneuron populations without increasing GABAergic synaptic density. Postnatal E2 treatment may induce lasting transcriptional changes that lead to enduring disease modification and could potentially serve as a therapy for inherited interneuronopathies.
Collapse
Affiliation(s)
- Pedro R Olivetti
- Blue Bird Circle Developmental Neurogenetics Laboratory, Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | |
Collapse
|
44
|
Ball GF, Balthazart J, McCarthy MM. Is it useful to view the brain as a secondary sexual characteristic? Neurosci Biobehav Rev 2014; 46 Pt 4:628-38. [PMID: 25195165 DOI: 10.1016/j.neubiorev.2014.08.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 08/04/2014] [Accepted: 08/20/2014] [Indexed: 12/21/2022]
Abstract
Many sex differences in brain and behavior related to reproduction are thought to have evolved based on sexual selection involving direct competition for mates during male-male competition and female choice. Therefore, certain aspects of brain circuitry can be viewed as secondary sexual characteristics. The study of proximate causes reveals that sex differences in the brain of mammals and birds reflect organizational and activational effects of sex steroids as articulated by Young and collaborators. However, sex differences in brain and behavior have been identified in the cognitive domain with no obvious link to reproduction. Recent views of sexual selection advocate for a broader view of how intra-sexual selection might occur including such examples as competition within female populations for resources that facilitate access to mates rather than mating competition per se. Sex differences can also come about for other reasons than sexual selection and recent work on neuroendocrine mechanisms has identified a plethora of ways that the brain can develop in a sex specific manner. Identifying the brain as sexually selected requires careful hypothesis testing so that one can link a sex-biased aspect of a neural trait to a behavior that provides an advantage in a competitive mating situation.
Collapse
Affiliation(s)
- Gregory F Ball
- Department of Psychological and Brain Sciences, Johns Hopkins University, 3400 N, Charles Street, Baltimore, MD 21218, USA.
| | - Jacques Balthazart
- GIGA Neuroscience, University of Liege, 1 boulevard de l'Hôpital, 4000 Liege, Belgium.
| | - Margaret M McCarthy
- Department of Pharmacology, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21210, USA
| |
Collapse
|
45
|
Kiyokage E, Toida K, Suzuki-Yamamoto T, Ishimura K. Cellular localization of 5α-reductase in the rat cerebellum. J Chem Neuroanat 2014; 59-60:8-16. [DOI: 10.1016/j.jchemneu.2014.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 04/11/2014] [Accepted: 04/25/2014] [Indexed: 01/14/2023]
|
46
|
Cruz G, Foster W, Paredes A, Yi KD, Uzumcu M. Long-term effects of early-life exposure to environmental oestrogens on ovarian function: role of epigenetics. J Neuroendocrinol 2014; 26:613-24. [PMID: 25040227 PMCID: PMC4297924 DOI: 10.1111/jne.12181] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 06/22/2014] [Accepted: 07/15/2014] [Indexed: 12/14/2022]
Abstract
Oestrogens play an important role in development and function of the brain and reproductive tract. Accordingly, it is considered that developmental exposure to environmental oestrogens can disrupt neural and reproductive tract development, potentially resulting in long-term alterations in neurobehaviour and reproductive function. Many chemicals have been shown to have oestrogenic activity, whereas others affect oestrogen production and turnover, resulting in the disruption of oestrogen signalling pathways. However, these mechanisms and the concentrations required to induce these effects cannot account for the myriad adverse effects of environmental toxicants on oestrogen-sensitive target tissues. Hence, alternative mechanisms are assumed to underlie the adverse effects documented in experimental animal models and thus could be important to human health. In this review, the epigenetic regulation of gene expression is explored as a potential target of environmental toxicants including oestrogenic chemicals. We suggest that toxicant-induced changes in epigenetic signatures are important mechanisms underlying the disruption of ovarian follicular development. In addition, we discuss how exposure to environmental oestrogens during early life can alter gene expression through effects on epigenetic control potentially leading to permanent changes in ovarian physiology.
Collapse
Affiliation(s)
- Gonzalo Cruz
- Centro de Neurobiología y Plasticidad Cerebral, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Correspondence to: Gonzalo Cruz, Gran Bretaña 1111, Playa Ancha, Valparaíso, Chile. 2360102, Tel. 56 32 2508015,
| | - Warren Foster
- Department of Obstetrics & Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Alfonso Paredes
- Laboratorio de Neurobioquímica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Chile
| | - Kun Don Yi
- Syngenta Crop Protection, LLC. Greensboro, NC
| | - Mehmet Uzumcu
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
47
|
Changes in memory and synaptic plasticity induced in male rats after maternal exposure to bisphenol A. Toxicology 2014; 322:51-60. [DOI: 10.1016/j.tox.2014.05.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 04/30/2014] [Accepted: 05/01/2014] [Indexed: 11/17/2022]
|
48
|
Ferri SL, Hildebrand PF, Way SE, Flanagan-Cato LM. Estradiol regulates markers of synaptic plasticity in the hypothalamic ventromedial nucleus and amygdala of female rats. Horm Behav 2014; 66:409-20. [PMID: 24995468 DOI: 10.1016/j.yhbeh.2014.06.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 06/17/2014] [Accepted: 06/24/2014] [Indexed: 01/28/2023]
Abstract
Ovarian hormones act in multiple brain regions to modulate specific behaviors and emotional states. For example, ovarian hormones promote female sexual receptivity in the hypothalamic ventromedial nucleus (VMH) and modulate anxiety in the amygdala. Hormone-induced changes within the VMH include structural modifications, such as changes in dendritic spines, dendrite length and the number of synapses. In some situations, dendrite remodeling requires actin polymerization, which depends on phospho-deactivation of the enzyme cofilin, or the ionotropic AMPA-type glutamate receptors, especially the GluA1 and GluA2 subunits. The present experiments used immunohistochemistry to test the hypothesis that ovarian hormone-induced neural plasticity in the VMH and amygdala involves the regulation of phospho-cofilin, GluA1 and GluA2. These proteins were assessed acutely after estradiol administration (0.5, 1.0 and 4.0h), as well as three days after hormone treatment. Both brain regions displayed rapid (4.0h or less) and transient estradiol-induced increases in the level of phospho-cofilin. At the behaviorally relevant time point of three days, differential changes in AMPA receptor subunits were observed. Using Golgi impregnation, the effect of estradiol on amygdala dendrites was examined. Three days after estradiol treatment, an increase in the length of dendrites in the central nucleus of the amygdala was observed. Thus, estradiol initiates structural changes in dendrites in both the VMH and amygdala associated with an early phospho-deactivation of cofilin, followed by dynamic, brain region-specific changes in AMPA receptor composition.
Collapse
Affiliation(s)
- Sarah L Ferri
- Department of Psychology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Peter F Hildebrand
- Department of Psychology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Samantha E Way
- Department of Psychology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Loretta M Flanagan-Cato
- Department of Psychology, University of Pennsylvania, Philadelphia, PA 19104, USA; Mahoney Institute of Neurological Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
49
|
Bian C, Zhu H, Zhao Y, Cai W, Zhang J. Intriguing roles of hippocampus-synthesized 17β-estradiol in the modulation of hippocampal synaptic plasticity. J Mol Neurosci 2014; 54:271-81. [PMID: 24729128 DOI: 10.1007/s12031-014-0285-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/06/2014] [Indexed: 10/25/2022]
Abstract
Accumulated studies have shown that 17β-estradiol (E2) can be de novo synthesized in the hippocampus, and its role in the regulation of hippocampal synaptic plasticity, which is the basis of learning and memory, has long been exploring. Steroidogenic enzymes (e.g., aromatase) that are essential to the hippocampus-synthesized synthesis of E2 have been detected in the hippocampus. Inhibition of E2 synthesis by aromatase inhibitors significantly reduces the density of hippocampal spine synapses, levels of some synaptic proteins such as spinopholin and synaptophysin. Moreover, the electrophysiological properties of hippocampal neurons are also changed in response to this inhibition. The influences of gonadal and hippocampal E2 on synaptic plasticity may exist some differences, since some reports showed that gonadal (or circulating) estrogens have no obvious effects in the modulation of hippocampal synaptic proteins as evidenced in some ovariectomized animals and postmenopausal women who suffered from Alzheimer's disease (AD). These evidences leads to a hypothesis that hippocampal E2 may play a more important role in modulation of synaptic plasticity than gonadal E2. The signaling pathways, whereby hippocampal E2 modulates synaptic plasticity, insist of classical chronic genomic pathway and rapid nongenomic pathway, which mediated by nonnuclear estrogen receptor (GPER) and/or nuclear or nonnuclear estrogen receptors, which require coactivators for their transcription activity. Among which steroid receptor coactivator-1 (SRC-1) is the predominant coactivator p160 family members in the brain. Several clues have shown that SRC-1 is expressed in hippocampus and is highly correlated with some key synaptic proteins developmentally or after orchidectomy but not ovariectomy, indicating SRC-1 may be regulated by hippocampus-synthesized E2 and profoundly involved in the mediation of hippocampal E2 regulation of hippocampal synaptic plasticity. Further studies about the exact roles of hippocampus-synthesized E2 and therefore SRC-1 are urgently needed in order to facilitate our understanding of hippocampal E2, which will be very important to the development of novel strategies of estrogen replacement therapy against neurodegenerative deficits such as Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Chen Bian
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Gaotanyan 30, Chongqing, 400038, China
| | | | | | | | | |
Collapse
|
50
|
Hart D, Nilges M, Pollard K, Lynn T, Patsos O, Shiel C, Clark SM, Vasudevan N. Activation of the G-protein coupled receptor 30 (GPR30) has different effects on anxiety in male and female mice. Steroids 2014; 81:49-56. [PMID: 24240011 DOI: 10.1016/j.steroids.2013.11.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The GPR30, a former orphan GPCR, is a putative membrane estrogen receptor that can activate rapid signaling pathways such as extracellular regulated kinase (ERK) in a variety of cells and may contribute to estrogen's effects in the central nervous system. The distribution of GPR30 in the limbic system predicts a role for this receptor in the regulation of learning and memory and anxiety by estrogens. Though acute G-1 treatment is reported to be anxiogenic in ovariectomised female mice and in gonadally intact male mice, the effect of GPR30 activation is unknown in gonadectomised male mice. In this study, we show that an acute administration of G-1 to gonadectomised male mice, but not female mice, was anxiolytic on an elevated plus maze task, without affecting locomotor activity. In addition, though G-1 treatment did not regulate ERK, it was associated with increased estrogen receptor (ER)α phosphorylation in the ventral, but not dorsal, hippocampus of males. In the female, G-1 increased the ERK activation solely in the dorsal hippocampus, independent of state anxiety. This is the first study to report an anxiolytic effect of GPR30 activation in male mice, in a rapid time frame that is commensurate with non-genomic signaling by estrogen.
Collapse
Affiliation(s)
- David Hart
- Biological Chemistry Program, Tulane University, New Orleans, LA 70118, United States
| | - Mary Nilges
- Neuroscience Program, Tulane University, New Orleans, LA 70118, United States
| | - Kevin Pollard
- Neuroscience Program, Tulane University, New Orleans, LA 70118, United States
| | - Tucker Lynn
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, United States
| | - Olivia Patsos
- Neuroscience Program, Tulane University, New Orleans, LA 70118, United States
| | - Cassidy Shiel
- Department of Psychology, Tulane University, New Orleans, LA 70118, United States
| | - Sara M Clark
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, United States
| | - Nandini Vasudevan
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, United States; Neuroscience Program, Tulane University, New Orleans, LA 70118, United States.
| |
Collapse
|