1
|
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: 2.0] [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
|
2
|
Garcia RG, Staley R, Aroner S, Stowell J, Sclocco R, Napadow V, Barbieri R, Goldstein JM. Optimization of respiratory-gated auricular vagus afferent nerve stimulation for the modulation of blood pressure in hypertension. Front Neurosci 2022; 16:1038339. [PMID: 36570845 PMCID: PMC9783922 DOI: 10.3389/fnins.2022.1038339] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/14/2022] [Indexed: 12/13/2022] Open
Abstract
Background The objective of this pilot study was to identify frequency-dependent effects of respiratory-gated auricular vagus afferent nerve stimulation (RAVANS) on the regulation of blood pressure and heart rate variability in hypertensive subjects and examine potential differential effects by sex/gender or race. Methods Twenty hypertensive subjects (54.55 ± 6.23 years of age; 12 females and 8 males) were included in a within-person experimental design and underwent five stimulation sessions where they received RAVANS at different frequencies (i.e., 2 Hz, 10 Hz, 25 Hz, 100 Hz, or sham stimulation) in a randomized order. EKG and continuous blood pressure signals were collected during a 10-min baseline, 30-min stimulation, and 10-min post-stimulation periods. Generalized estimating equations (GEE) adjusted for baseline measures were used to evaluate frequency-dependent effects of RAVANS on heart rate, high frequency power, and blood pressure measures, including analyses stratified by sex and race. Results Administration of RAVANS at 100 Hz had significant overall effects on the reduction of heart rate (β = -2.03, p = 0.002). It was also associated with a significant reduction of diastolic (β = -1.90, p = 0.01) and mean arterial blood pressure (β = -2.23, p = 0.002) in Black hypertensive participants and heart rate in female subjects (β = -2.83, p = 0.01) during the post-stimulation period when compared to sham. Conclusion Respiratory-gated auricular vagus afferent nerve stimulation exhibits frequency-dependent rapid effects on the modulation of heart rate and blood pressure in hypertensive patients that may further differ by race and sex. Our findings highlight the need for the development of optimized stimulation protocols that achieve the greatest effects on the modulation of physiological and clinical outcomes in this population.
Collapse
Affiliation(s)
- Ronald G. Garcia
- Clinical Neuroscience Laboratory of Sex Differences in the Brain, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- School of Medicine, Universidad de Santander, Bucaramanga, Colombia
| | - Rachel Staley
- Clinical Neuroscience Laboratory of Sex Differences in the Brain, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sarah Aroner
- Clinical Neuroscience Laboratory of Sex Differences in the Brain, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Jessica Stowell
- Clinical Neuroscience Laboratory of Sex Differences in the Brain, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Roberta Sclocco
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Scott Schoen and Nancy Adams Discovery Center for Recovery from Chronic Pain, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, United States
- Department of Gastroenterology and Center for Neurointestinal Health, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Vitaly Napadow
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Scott Schoen and Nancy Adams Discovery Center for Recovery from Chronic Pain, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, United States
| | - Riccardo Barbieri
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
- Department of Anesthesia and Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Jill M. Goldstein
- Clinical Neuroscience Laboratory of Sex Differences in the Brain, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
3
|
Long M, Puhlmann L, Vrtička P. Hypothalamus volume in men: Investigating associations with paternal status, self-reported caregiving beliefs, and adult attachment style. Soc Neurosci 2021; 16:639-652. [PMID: 34704890 DOI: 10.1080/17470919.2021.1997799] [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] [Indexed: 10/20/2022]
Abstract
Most studies on mammalian caregiving and attachment focused on the mother-child relationship, particularly in humans. Yet, changing societal roles of male caregivers have highlighted the necessity for research with fathers.We examined the volume of the hypothalamus, an important subcortical brain area for caregiving and attachment, in N = 50 fathering (child age 5-6 years) and N = 45 non-fathering men using a novel technique to identify the hypothalamus in 3T MRI. We furthermore employed three self-report measures to assess interindividual differences in adult attachment style across all men and caregiving beliefs in fathers.While we did not observe any significant difference in hypothalamus volume between fathers and non-fathers or associations between hypothalamus volume and self-reported adult attachment style across all men, self-reported caregiving beliefs were positively related to total hypothalamus volume in fathers. A follow-up analysis showed that fathers' self-reported belief that a father's role is important to child development was specifically related to tuberal hypothalamus volume, while self-reported enjoyment of spending time with the child was not associated with sub-regional hypothalamus volume.Together, these findings suggest that interindividual variability in self-reported caregiving beliefs in fathers is related to brain structure, warranting further research.
Collapse
Affiliation(s)
- M Long
- Alberta Children's Hospital Research Institute, University of Calgary, Canada.,Research Group "Social Stress and Family Health", Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - L Puhlmann
- Research Group "Social Stress and Family Health", Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.,Leibniz Institute for Resilience Research (LIR), Mainz, Germany
| | - P Vrtička
- Research Group "Social Stress and Family Health", Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.,Centre for Brain Science, Department of Psychology, University of Essex, Colchester, UK
| |
Collapse
|
4
|
Mareckova K, Marecek R, Andryskova L, Brazdil M, Nikolova YS. Impact of prenatal stress on amygdala anatomy in young adulthood: Timing and location matter. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 7:231-238. [PMID: 34358683 DOI: 10.1016/j.bpsc.2021.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Exposure to maternal stress in utero has long-term implications for the developing brain and has been linked with a higher risk of depression. The amygdala, which develops during the early embryonic stage and is critical for emotion processing, might be particularly sensitive. METHODS Using data from a neuroimaging follow-up of the ELSPAC prenatal birth cohort (n=129, 47% men, 23-24 years old), we studied the impact of prenatal stress during the first and second half of pregnancy on the volume of the amygdala and its nuclei in young adult offspring. We further evaluated the relationship between amygdala anatomy and offspring depressive symptomatology. Amygdala nuclei were parcellated using FreeSurfer's automated segmentation pipeline. Depressive symptoms were measured via self-report using the Beck Depression Inventory (BDI). RESULTS Exposure to stress during the first half of pregnancy was associated with smaller accessory basal (Cohen's f2=0.27, p(FDR)=0.03) and cortical (Cohen's f2=0.29, p(FDR)=0.03) nuclei volumes. This effect remained significant after correcting for sex, stress during the second half of pregnancy, as well as maternal age at birth, birth weight, maternal education, and offspring's age at MRI. These two nuclei showed a quadratic relationship with BDI scores in young adulthood, where both smaller and larger volume was associated with more depressive symptoms (Accessory basal nucleus: Adj R2=0.05. p(FDR)=0.015; Cortical nucleus: Adj R2=0.04, p(FDR)=0.015). CONCLUSIONS We conclude that exposure to stress during the first half of pregnancy might have long-term implications for amygdala anatomy, which may in turn predict the experience of depressive symptoms in young adulthood.
Collapse
Affiliation(s)
- Klara Mareckova
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Brain and Mind Research, Central European Institute of Technology, Masaryk University, Brno, Czech Republic.
| | - Radek Marecek
- Brain and Mind Research, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Lenka Andryskova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Milan Brazdil
- Brain and Mind Research, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Yuliya S Nikolova
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
5
|
Impact of prenatal maternal cytokine exposure on sex differences in brain circuitry regulating stress in offspring 45 years later. Proc Natl Acad Sci U S A 2021; 118:2014464118. [PMID: 33876747 DOI: 10.1073/pnas.2014464118] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Stress is associated with numerous chronic diseases, beginning in fetal development with in utero exposures (prenatal stress) impacting offspring's risk for disorders later in life. In previous studies, we demonstrated adverse maternal in utero immune activity on sex differences in offspring neurodevelopment at age seven and adult risk for major depression and psychoses. Here, we hypothesized that in utero exposure to maternal proinflammatory cytokines has sex-dependent effects on specific brain circuitry regulating stress and immune function in the offspring that are retained across the lifespan. Using a unique prenatal cohort, we tested this hypothesis in 80 adult offspring, equally divided by sex, followed from in utero development to midlife. Functional MRI results showed that exposure to proinflammatory cytokines in utero was significantly associated with sex differences in brain activity and connectivity during response to negative stressful stimuli 45 y later. Lower maternal TNF-α levels were significantly associated with higher hypothalamic activity in both sexes and higher functional connectivity between hypothalamus and anterior cingulate only in men. Higher prenatal levels of IL-6 were significantly associated with higher hippocampal activity in women alone. When examined in relation to the anti-inflammatory effects of IL-10, the ratio TNF-α:IL-10 was associated with sex-dependent effects on hippocampal activity and functional connectivity with the hypothalamus. Collectively, results suggested that adverse levels of maternal in utero proinflammatory cytokines and the balance of pro- to anti-inflammatory cytokines impact brain development of offspring in a sexually dimorphic manner that persists across the lifespan.
Collapse
|
6
|
Ge R, Liu X, Long D, Frangou S, Vila-Rodriguez F. Sex effects on cortical morphological networks in healthy young adults. Neuroimage 2021; 233:117945. [PMID: 33711482 DOI: 10.1016/j.neuroimage.2021.117945] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/17/2021] [Accepted: 03/03/2021] [Indexed: 12/30/2022] Open
Abstract
Understanding sex-related differences across the human cerebral cortex is an important step in elucidating the basis of psychological, behavioural and clinical differences between the sexes. Prior structural neuroimaging studies primarily focused on regional sex differences using univariate analyses. Here we focus on sex differences in cortical morphological networks (CMNs) derived using multivariate modelling of regional cortical measures of volume and surface from high-quality structural MRI scans from healthy participants in the Human Connectome Project (HCP) (n = 1,063) and the Southwest University Longitudinal Imaging Multimodal (SLIM) study (n = 549). The functional relevance of the CMNs was inferred using the NeuroSynth decoding function. Sex differences were widespread but not uniform. In general, females had higher volume, thickness and cortical folding in networks that involve prefrontal (both ventral and dorsal regions including the anterior cingulate) and parietal regions while males had higher volume, thickness and cortical folding in networks that primarily include temporal and posterior cortical regions. CMN loading coefficients were used as input features to linear discriminant analyses that were performed separately in the HCP and SLIM; sex was predicted with a high degree of accuracy (81%-85%) across datasets. The availability of behavioral data in the HCP enabled us to show that male-biased surface-based CMNs were associated with externalizing behaviors. These results extend previous literature on regional sex-differences by identifying CMNs that can reliably predict sex, are relevant to the expression of psychopathology and provide the foundation for the future investigation of their functional significance in clinical populations.
Collapse
Affiliation(s)
- Ruiyang Ge
- Non-Invasive Neurostimulation Therapies Laboratory, Department of Psychiatry, University of British Columbia, BC, Canada
| | - Xiang Liu
- Non-Invasive Neurostimulation Therapies Laboratory, Department of Psychiatry, University of British Columbia, BC, Canada
| | - David Long
- Non-Invasive Neurostimulation Therapies Laboratory, Department of Psychiatry, University of British Columbia, BC, Canada
| | - Sophia Frangou
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, BC, Canada; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Fidel Vila-Rodriguez
- Non-Invasive Neurostimulation Therapies Laboratory, Department of Psychiatry, University of British Columbia, BC, Canada.
| |
Collapse
|
7
|
Impact of sex and depressed mood on the central regulation of cardiac autonomic function. Neuropsychopharmacology 2020; 45:1280-1288. [PMID: 32152473 PMCID: PMC7298013 DOI: 10.1038/s41386-020-0651-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 02/21/2020] [Accepted: 02/28/2020] [Indexed: 12/31/2022]
Abstract
Cardiac autonomic dysregulation has been implicated in the comorbidity of major psychiatric disorders and cardiovascular disease, potentially through dysregulation of physiological responses to negative stressful stimuli (here, shortened to stress response). Further, sex differences in these comorbidities are substantial. Here, we tested the hypothesis that mood- and sex-dependent alterations in brain circuitry implicated in the regulation of the stress response are associated with reduced peripheral parasympathetic activity during negative emotional arousal. Fifty subjects (28 females) including healthy controls and individuals with major depression, bipolar psychosis and schizophrenia were evaluated. Functional magnetic resonance imaging and physiology (cardiac pulse) data were acquired during a mild visual stress reactivity challenge. Associations between changes in activity and functional connectivity of the stress response circuitry and variations in cardiovagal activity [normalized high frequency power of heart rate variability (HFn)] were evaluated using GLM analyses, including interactions with depressed mood and sex across disorders. Our results revealed that in women with high depressed mood, lower cardiovagal activity in response to negative affective stimuli was associated with greater activation of hypothalamus and right amygdala and reduced connectivity between hypothalamus and right orbitofrontal cortex, amygdala, and hippocampus. No significant associations were observed in women with low levels of depressed mood or men. Our results revealed mood- and sex-dependent interactions in the central regulation of cardiac autonomic activity in response to negative affective stimuli. These findings provide a potential pathophysiological mechanism for previously observed sex differences in the comorbidity of major depression and cardiovascular disease.
Collapse
|
8
|
Bi D, Wen L, Wu Z, Shen Y. GABAergic dysfunction in excitatory and inhibitory (E/I) imbalance drives the pathogenesis of Alzheimer's disease. Alzheimers Dement 2020; 16:1312-1329. [PMID: 32543726 DOI: 10.1002/alz.12088] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/28/2020] [Accepted: 02/10/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To propose a new hypothesis that GABAergic dysfunction in excitatory and inhibitory (E/I) imbalance drives the pathogenesis of Alzheimer's disease (AD). BACKGROUND Synaptic dysfunction and E/I imbalance emerge decades before the appearance of cognitive decline in AD patients, which contribute to neurodegeneration. Initially, E/I imbalance was thought to occur first, due to dysfunction of the glutamatergic and cholinergic systems. However, new evidence has demonstrated that the GABAergic system, the counterpart of E/I balance and the major inhibitory neurotransmitter system in the central nervous system, is altered enormously and that this contributes to E/I imbalance and further AD pathogenesis. NEW HYPOTHESIS Alterations to the GABAergic system, induced by multiple AD pathogenic or risk factors, contribute to E/I imbalance and AD pathogenesis. MAJOR CHALLENGES FOR THE HYPOTHESIS This GABAergic hypothesis accounts for many critical questions and common challenges confronting a new hypothesis of AD pathogenesis. More specifically, it explains why amyloid beta (Aβ), β-secretase (BACE1), apolipoprotein E4 gene (APOE ε4), hyperactive glia cells, contributes to AD pathogenesis and why age and sex are the risk factors of AD. GABAergic dysfunction promotes the spread of Aβ pathology throughout the AD brain and associated cognitive impairments, and the induction of dysfunction induced by these varied risk factors shares this common neurobiology leading to E/I imbalance. In turn, some of these factors exacerbate GABAergic dysfunction and E/I imbalance. Moreover, the GABAergic system modulates various brain functions and thus, the GABAergic hypothesis accounts for nonamnestic manifestations. Furthermore, corrections of E/I balance through manipulation of GABAergic functions have shown positive outcomes in preclinical and clinical studies, suggesting the potential of the GABAergic system as a therapeutic target in AD. LINKAGE TO OTHER MAJOR THEORIES Dysfunction of the GABAergic system is induced by multiple critical signaling pathways, which include the existing major theories of AD pathogenesis, such as the Aβ and neuroinflammation hypotheses. In a new perspective, this GABAergic hypothesis accounts for the E/I imbalance and related excitotoxicity, which contribute to cognitive decline and AD pathogenesis. Therefore, the GABAergic system could be a key target to restore, at least partially, the E/I balance and cognitive function in AD patients.
Collapse
Affiliation(s)
- Danlei Bi
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, University of Sciences and Technology of China, Hefei, China.,Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lang Wen
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, University of Sciences and Technology of China, Hefei, China.,Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zujun Wu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, University of Sciences and Technology of China, Hefei, China.,Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yong Shen
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, University of Sciences and Technology of China, Hefei, China.,Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
9
|
Bizzozzero-Hiriart M, Di Giorgio NP, Libertun C, Lux-Lantos V. GABAergic input through GABA B receptors is necessary during a perinatal window to shape gene expression of factors critical to reproduction such as Kiss1. Am J Physiol Endocrinol Metab 2020; 318:E901-E919. [PMID: 32286880 DOI: 10.1152/ajpendo.00547.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Lack of GABAB receptors in GABAB1 knockout mice decreases neonatal ARC kisspeptin 1 (Kiss1) expression in the arcuate nucleus of the hypothalamus (ARC) in females, which show impaired reproduction as adults. Our aim was to selectively impair GABAB signaling during a short postnatal period to evaluate its impact on the reproductive system. Neonatal male and female mice were injected with the GABAB antagonist CGP 55845 (CGP, 1 mg/kg body wt sc) or saline from postnatal day 2 (PND2) to PND6, three times per day (8 AM, 1 PM, and 6 PM). One group was killed on PND6 for collection of blood samples (hormones by radioimmunoassay), brains for gene expression in the anteroventral periventricular nucleus-periventricular nucleus continuum (AVPV/PeN), and ARC micropunches [quantitative PCR (qPCR)] and gonads for qPCR, hormone contents, and histology. A second group of mice was injected with CGP (1 mg/kg body wt sc) or saline from PND2 to PND6, three times per day (8 AM, 1 PM, and 6 PM), and left to grow to adulthood. We measured body weight during development and parameters of sexual differentiation, puberty onset, and estrous cycles. Adult mice were killed, and trunk blood (hormones), brains for qPCR, and gonads for qPCR and hormone contents were obtained. Our most important findings on PND6 include the CGP-induced decrease in ARC Kiss1 and increase in neurokinin B (Tac2) in both sexes; the decrease in AVPV/PeN tyrosine hydroxylase (Th) only in females; the increase in gonad estradiol content in both sexes; and the increase in primordial follicles and decrease in primary and secondary follicles. Neonatally CGP-treated adults showed decreased ARC Kiss1 and ARC gonadotropin-releasing hormone (Gnrh1) and increased ARC glutamic acid decarboxylase 67 (Gad1) only in males; increased ARC GABAB receptor subunit 1 (Gabbr1) in both sexes; and decreased AVPV/PeN Th only in females. We demonstrate that ARC Kiss1 expression is chronically downregulated in males and that the normal sex difference in AVPV/PeN Th expression is abolished. In conclusion, neonatal GABAergic input through GABAB receptors shapes gene expression of factors critical to reproduction.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Arcuate Nucleus of Hypothalamus/drug effects
- Arcuate Nucleus of Hypothalamus/metabolism
- Estradiol/metabolism
- Female
- Follicle Stimulating Hormone/metabolism
- GABA-B Receptor Antagonists/pharmacology
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/physiology
- Glutamate Decarboxylase/genetics
- Glutamate Decarboxylase/metabolism
- Gonadotropin-Releasing Hormone/genetics
- Gonadotropin-Releasing Hormone/metabolism
- Hypothalamus, Anterior/drug effects
- Hypothalamus, Anterior/metabolism
- Kisspeptins/genetics
- Kisspeptins/metabolism
- Luteinizing Hormone/metabolism
- Male
- Mice
- Ovary/drug effects
- Ovary/metabolism
- Phosphinic Acids/pharmacology
- Propanolamines/pharmacology
- Protein Precursors/genetics
- Protein Precursors/metabolism
- Puberty/drug effects
- Puberty/genetics
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Receptors, GABA-B/genetics
- Receptors, GABA-B/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Reproduction/drug effects
- Reproduction/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Sex Differentiation/drug effects
- Sex Differentiation/genetics
- Tachykinins/genetics
- Tachykinins/metabolism
- Testis/drug effects
- Testis/metabolism
- Testosterone/metabolism
- Tyrosine 3-Monooxygenase/genetics
- Tyrosine 3-Monooxygenase/metabolism
Collapse
Affiliation(s)
- Marianne Bizzozzero-Hiriart
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Noelia P Di Giorgio
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Carlos Libertun
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Victoria Lux-Lantos
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| |
Collapse
|
10
|
de Matos LO, de Araujo Lima Reis AL, Lopes Guerra LT, de Oliveira Guarnieri L, Moraes MA, Arabe LB, de Souza RP, Pereira GS, Souza BR. Early postnatal l-Dopa treatment causes behavioral alterations in female vs. male young adult Swiss mice. Neuropharmacology 2020; 170:108047. [DOI: 10.1016/j.neuropharm.2020.108047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/28/2020] [Accepted: 03/08/2020] [Indexed: 01/21/2023]
|
11
|
Di Giorgio NP, Bizzozzero-Hiriart M, Libertun C, Lux-Lantos V. Unraveling the connection between GABA and kisspeptin in the control of reproduction. Reproduction 2020; 157:R225-R233. [PMID: 30844750 DOI: 10.1530/rep-18-0527] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 03/07/2019] [Indexed: 12/20/2022]
Abstract
Neuroendocrine control of reproduction involves the interplay of various factors that become active at some point along development. GnRH is the main neurohormone controlling reproduction and among the most important inputs modulating GnRH synthesis/secretion are GABA and kisspeptins. These interactions of GABA and kisspeptin in the control of GnRH secretion can take place by the presence of the receptors of both factors on the GnRH neuron or alternatively by the actions of GABA on kisspeptin neurons and/or the actions of kisspeptin on GABA neurons. Kisspeptin acts on the Kiss1R, a seven transmembrane domain, Gαq/11-coupled receptor that activates phospholipase C, although some Gαq/11-independent pathways in mediating part of the effects of Kiss1R activation have also been proposed. GABA acts through two kinds of receptors, ionotropic GABAA/C receptors involving a chloride channel and associated with fast inhibitory/stimulatory conductance and metabotropic GABAB receptors (GABABR) that are Gi/0 protein linked inducing late slow hyperpolarization. In this review, we aim to summarize the different ways in which these two actors, kisspeptin and GABA, interact to modulate GnRH secretion across the reproductive lifespan.
Collapse
Affiliation(s)
- Noelia P Di Giorgio
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | | | - Carlos Libertun
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina.,Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Victoria Lux-Lantos
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| |
Collapse
|
12
|
Impact of quercetin on tight junctional proteins and BDNF signaling molecules in hippocampus of PCBs-exposed rats. Interdiscip Toxicol 2019; 11:294-305. [PMID: 31762681 PMCID: PMC6853011 DOI: 10.2478/intox-2018-0029] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 02/06/2018] [Indexed: 12/26/2022] Open
Abstract
Polychlorinated biphenyls (PCBs) consist of a range of toxic substances which are directly proportional to carcinogenesis and tumor-promoting factors as well as having neurotoxic properties. Reactive oxygen species, which are produced from PCBs, alter blood–brain barrier (BBB) integrity, which is paralleled by cytoskeletal rearrangements and redistribution and disappearance of tight junction proteins (TJPs) like claudin-5 and occludin. Brain-derived neurotrophic factor (BDNF), plays an important role in the maintenance, survival of neurons and synaptic plasticity. It is predominant in the hippocampal areas vital to learning, memory and higher thinking. Quercetin, a flavonoid, had drawn attention to its neurodefensive property. The study is to assess the role of quercetin on serum PCB, estradiol and testosterone levels and mRNA expressions of estrogen receptor α and β, TJPs and BDNF signaling molecules on the hippocampus of PCBs-exposed rats. Rats were divided into 4 groups of 6 each. Group I rats were intraperitoneally (i.p.) administered corn oil (vehicle). Group II received quercetin 50 mg/kg/bwt (gavage). Group III received PCBs (Aroclor 1254) at 2 mg/kg bwt (i.p). Group IV received quercetin 50 mg/kg bwt (gavage) simultaneously with PCBs 2 mg/kg bwt (i.p.). The treatment was given daily for 30 days. The rats were euthanized 24 h after the experimental period. Blood was collected for quantification of serum PCBs estradiol and testosterone. The hippocampus was dissected and processed for PCR and Western blot; serum PCB was observed in PCB treated animals, simultaneously quercetin treated animals showed PCB metabolites. Serum testosterone and estradiol were decreased after PCB exposure. Quercetin supplementation brought back normal levels. mRNA expressions of estrogen α and β were decreased in the hippocampus of PCB treated rats. TJPS and BDNF signalling molecules were decreased in hippocampus of PCB treated rats. Quercetin supplementation retrieved all the parameters. Quercetin alone treated animals showed no alteration. Thus in PCB caused neurotoxicity, quercetin protects and prevents neuronal damage in the hippocampus.
Collapse
|
13
|
CHEN G, WANG X, TANG D. [Progress on evaluation, diagnosis and management of disorders of sex development]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2019; 48:358-366. [PMID: 31901037 PMCID: PMC8800683 DOI: 10.3785/j.issn.1008-9292.2019.08.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/30/2019] [Indexed: 06/10/2023]
Abstract
Disorders of sex development (DSD) refer to a group of diseases characterized by abnormal congenital development of chromosomes, gonad or genitals with different pathophysiological changes and clinical manifestations. DSD is more common in neonates and adolescents, and neonates often show genital abnormalities while adolescents show abnormal sexual development during puberty. It is the international consensus that the scope of DSD should include basic clinical evaluation (internal and external genitalia and endocrine hormones), diagnostic confirmation (chromosome, genetic diagnosis), psychological assessment for children and family, treatment (sex assignment, hormone replacement and surgical intervention), potential fertility protection and long-term follow-up, which require the expertise of pediatric endocrinology, pediatric urology, clinical psychology, genetic disciplines, medical images and other related disciplines; that is, individualized management of children with DSD requires an experienced multidisciplinary team (MDT). This article reviews the recent progress on the evaluation, diagnosis and management of disorders of sex development.
Collapse
Affiliation(s)
| | | | - Daxing TANG
- 唐达星(1961—), 男, 博士, 硕士生导师, 主任医师, 主要从事小儿泌尿外科学研究; E-mail:
;
https://orcid.org/0000-0003-2103-4931
| |
Collapse
|
14
|
Goldstein JM, Hale T, Foster SL, Tobet SA, Handa RJ. Sex differences in major depression and comorbidity of cardiometabolic disorders: impact of prenatal stress and immune exposures. Neuropsychopharmacology 2019; 44:59-70. [PMID: 30030541 PMCID: PMC6235859 DOI: 10.1038/s41386-018-0146-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/15/2018] [Accepted: 06/21/2018] [Indexed: 12/11/2022]
Abstract
Major depressive disorder topped ischemic heart disease as the number one cause of disability worldwide in 2012, and women have twice the risk of men. Further, the comorbidity of depression and cardiometabolic disorders will be one of the primary causes of disability worldwide by 2020, with women at twice the risk. Thus, understanding the sex-dependent comorbidities has public health consequences worldwide. We propose here that sex differences in MDD-cardiometabolic comorbidity originate, in part, from pathogenic processes initiated in fetal development that involve sex differences in shared pathophysiology between the brain, the vascular system, the CNS control of the heart and associated hormonal, immune, and metabolic physiology. Pathways implicate neurotrophic and angiogenic growth factors, gonadal hormone receptors, and neurotransmitters such as gamma amino butyric acid (GABA) on neuronal and vascular development of HPA axis regions, such as the paraventricular nucleus (PVN), in addition to blood pressure, in part through the renin-angiotensin system, and insulin and glucose metabolism. We show that the same prenatal exposures have consequences for sex differences across multiple organ systems that, in part, share common pathophysiology. Thus, we believe that applying a sex differences lens to understanding shared biologic substrates underlying these comorbidities will provide novel insights into the development of sex-dependent therapeutics. Further, taking a lifespan perspective beginning in fetal development provides the opportunity to target abnormalities early in the natural history of these disorders in a sex-dependent way.
Collapse
Affiliation(s)
- Jill M Goldstein
- Departments of Psychiatry and Obstetrics and Gynecology, Massachusetts General Hospital (MGH), Boston, MA, 02120, USA.
- Departments of Psychiatry and Medicine, Harvard Medical School, Boston, MA, USA.
| | - Taben Hale
- Department of Basic Medical Science, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, 85004, USA
| | - Simmie L Foster
- Department of Psychiatry, Harvard Medical School, at Massachusetts General Hospital, Boston, MA, USA
| | - Stuart A Tobet
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| | - Robert J Handa
- Department of Basic Medical Science, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, 85004, USA
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| |
Collapse
|
15
|
de Matos LO, Reis ALDAL, Guerra LTL, Guarnieri LDO, Moraes MA, Aquino NSS, Szawka RE, Pereira GS, Souza BR. l-Dopa treatment during perinatal development leads to different behavioral alterations in female vs. male juvenile Swiss mice. Pharmacol Biochem Behav 2018; 173:1-14. [DOI: 10.1016/j.pbb.2018.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/25/2018] [Accepted: 08/06/2018] [Indexed: 12/26/2022]
|
16
|
Ali SA, Begum T, Reza F. Hormonal Influences on Cognitive Function. Malays J Med Sci 2018; 25:31-41. [PMID: 30914845 PMCID: PMC6422548 DOI: 10.21315/mjms2018.25.4.3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/10/2018] [Indexed: 11/09/2022] Open
Abstract
This article examines how hormonal changes may affect the neuronal networking and mechanisms of cognitive function. Hormones are the chemical regulators of the human body and function critically to maintain various processes, such as growth, emotions and even cognition. Numerous studies have examined the relationship between hormonal effects and cognitive function; these studies have investigated different factors, such as aging, pregnancy, post-natal states, emotions and stress. Different types of hormones produce different outcomes for the human body and mind. Hormones may also contribute to both positive and negative outcomes, depending on whether the hormone levels are too low or too high. To investigate the hormonal effects on cognitive function, the sources of localisation must be localised, so that the neuronal network can be realised. Furthermore, cognitive function does not rely on a specific brain region but is determined by the neuronal network interactions. Thus, it is worthwhile to know the neural mechanisms behind cognitive functions that are affected by hormones.
Collapse
Affiliation(s)
- Siti Atiyah Ali
- Department of Neurosciences, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Tahamina Begum
- Department of Neurosciences, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Faruque Reza
- Department of Neurosciences, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
17
|
Xie W, Cappiello M, Meng M, Rosenthal R, Zhang W. ADRA2B deletion variant and enhanced cognitive processing of emotional information: A meta-analytical review. Neurosci Biobehav Rev 2018; 92:402-416. [PMID: 29751052 DOI: 10.1016/j.neubiorev.2018.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/15/2018] [Accepted: 05/06/2018] [Indexed: 01/12/2023]
Abstract
This meta-analytical review examines whether a deletion variant in ADRA2B, a gene that encodes α2B adrenoceptor in the regulation of norepinephrine availability, influences cognitive processing of emotional information in human observers. Using a multilevel modeling approach, this meta-analysis of 16 published studies with a total of 2752 participants showed that ADRA2B deletion variant was significantly associated with enhanced perceptual and cognitive task performance for emotional stimuli. In contrast, this genetic effect did not manifest in overall task performance when non-emotional content was used. Furthermore, various study-level factors, such as targeted cognitive processes (memory vs. attention/perception) and task procedures (recall vs. recognition), could moderate the size of this genetic effect. Overall, with increased statistical power and standardized analytical procedures, this meta-analysis has established the contributions of ADRA2B to the interactions between emotion and cognition, adding to the growing literature on individual differences in attention, perception, and memory for emotional information in the general population.
Collapse
Affiliation(s)
- Weizhen Xie
- Department of Psychology, University of California, Riverside, United States.
| | - Marcus Cappiello
- Department of Psychology, University of California, Riverside, United States
| | - Ming Meng
- School of Psychology, South China Normal University, China
| | - Robert Rosenthal
- Department of Psychology, University of California, Riverside, United States
| | - Weiwei Zhang
- Department of Psychology, University of California, Riverside, United States
| |
Collapse
|
18
|
Mareckova K, Holsen L, Admon R, Whitfield-Gabrieli S, Seidman LJ, Buka SL, Klibanski A, Goldstein J. Neural - hormonal responses to negative affective stimuli: Impact of dysphoric mood and sex. J Affect Disord 2017; 222:88-97. [PMID: 28688266 PMCID: PMC5560420 DOI: 10.1016/j.jad.2017.06.050] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/07/2017] [Accepted: 06/22/2017] [Indexed: 01/15/2023]
Abstract
BACKGROUND Maladaptive responses to negative affective stimuli are pervasive, including clinically ill and healthy people, and men and women respond differently at neural and hormonal levels. Inspired by the Research Domain Criteria initiative, we used a transdiagnostic approach to investigate the impact of sex and dysphoric mood on neural-hormonal responses to negative affective stimuli. METHODS Participants included 99 individuals with major depressive disorder, psychosis and healthy controls. Functional magnetic resonance imaging (fMRI) was complemented with real-time acquisition of hypothalamo-pituitary-adrenal (HPA) and -gonadal (HPG) hormones. fMRI data were analyzed in SPM8 and task-related connectivity was assessed using generalized psychophysiological interaction. RESULTS Across all participants, elevated cortisol response predicted lower brain activity in orbitofrontal cortex and hypothalamus-amygdala connectivity. In those with worse dysphoric mood, elevated cortisol response predicted lower activity in hypothalamus and hippocampus. In women, elevated cortisol response was associated with lower activity in medial prefrontal cortex and low hypothalamo-hippocampal connectivity. In women with high dysphoric mood, elevated cortisol response was associated with low hypothalamo-hippocampal connectivity. There were no interactions with diagnosis or medication. LIMITATIONS There was limited power to correct for multiple comparisons across total number of ROIs and connectivity targets; cortisol responses were relatively low. CONCLUSIONS We conclude that the pathophysiology in neural-hormonal responses to negative affective stimuli is shared across healthy and clinical populations and varies as a function of sex and dysphoric mood. Our findings may contribute to the development of hormonal adjunctive therapeutics that are sex-dependent, underscoring the importance of one's sex to precision medicine.
Collapse
Affiliation(s)
- K. Mareckova
- Connors Center for Women’s Health and Gender Biology, Department of Medicine, Brigham and Women’s Hospital; Department of Psychiatry, Harvard Medical School (HMS), Boston, MA, USA,CEITEC, Masaryk University, Brno, Czech Republic
| | - L. Holsen
- Connors Center for Women’s Health and Gender Biology, Department of Medicine, Brigham and Women’s Hospital; Department of Psychiatry, Harvard Medical School (HMS), Boston, MA, USA
| | - R. Admon
- McLean Hospital, Department of Psychiatry, HMS, Boston, MA USA
| | - S. Whitfield-Gabrieli
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA
| | - LJ Seidman
- Beth Israel Deaconess Medical Center, Division of Public Psychiatry, Massachusetts Mental Health Center; Department of Psychiatry, HMS, Boston, MA, USA
| | - SL Buka
- Department of Community Health, Brown University, Providence, RI, USA
| | - A. Klibanski
- Massachusetts General Hospital, Department of Medicine, Neuroendocrine Unit; HMS, Department of Medicine, Boston, MA, USA
| | - J.M. Goldstein
- Connors Center for Women’s Health and Gender Biology, Department of Medicine, Brigham and Women’s Hospital; Department of Psychiatry, Harvard Medical School (HMS), Boston, MA, USA,Departments of Psychiatry and Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Goldstein JM, Holsen L, Huang G, Hammond BD, James-Todd T, Cherkerzian S, Hale TM, Handa RJ. Prenatal stress-immune programming of sex differences in comorbidity of depression and obesity/metabolic syndrome. DIALOGUES IN CLINICAL NEUROSCIENCE 2017. [PMID: 28179814 PMCID: PMC5286728 DOI: 10.31887/dcns.2016.18.4/jgoldstein] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Major depressive disorder (MDD) is the number one cause of disability worldwide and is comorbid with many chronic diseases, including obesity/metabolic syndrome (MetS). Women have twice as much risk for MDD and comorbidity with obesity/MetS as men, although pathways for understanding this association remain unclear. On the basis of clinical and preclinical studies, we argue that prenatal maternal stress (ie, excess glucocorticoid expression and associated immune responses) that occurs during the sexual differentiation of the fetal brain has sex-dependent effects on brain development within highly sexually dimorphic regions that regulate mood, stress, metabolic function, the autonomic nervous system, and the vasculature. Furthermore, these effects have lifelong consequences for shared sex-dependent risk of MDD and obesity/MetS. Thus, we propose that there are shared biologic substrates at the anatomical, molecular, and/or genetic levels that produce the comorbid risk for MDD-MetS through sex-dependent fetal origins.
Collapse
Affiliation(s)
- Jill M Goldstein
- Connors Center for Women's Health and Gender Biology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Departments of Psychiatry and Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Laura Holsen
- Connors Center for Women's Health and Gender Biology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Departments of Psychiatry and Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Grace Huang
- Division of Endocrinology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Bradley D Hammond
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA; Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Tamarra James-Todd
- Connors Center for Women's Health and Gender Biology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Sara Cherkerzian
- Connors Center for Women's Health and Gender Biology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Departments of Psychiatry and Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Taben M Hale
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA
| | - Robert J Handa
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA; Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
20
|
Chachlaki K, Garthwaite J, Prevot V. The gentle art of saying NO: how nitric oxide gets things done in the hypothalamus. Nat Rev Endocrinol 2017. [PMID: 28621341 DOI: 10.1038/nrendo.2017.69] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The chemical signalling molecule nitric oxide (NO), which freely diffuses through aqueous and lipid environments, subserves an array of functions in the mammalian central nervous system, such as the regulation of synaptic plasticity, blood flow and neurohormone secretion. In this Review, we consider the cellular and molecular mechanisms by which NO evokes short-term and long-term changes in neuronal activity. We also highlight recent studies showing that discrete populations of neurons that synthesize NO in the hypothalamus constitute integrative systems that support life by relaying metabolic and gonadal signals to the neuroendocrine brain, and thus gate the onset of puberty and adult fertility. The putative involvement and therapeutic potential of NO in the pathophysiology of brain diseases, for which hormonal imbalances during postnatal development could be risk factors, is also discussed.
Collapse
Affiliation(s)
- Konstantina Chachlaki
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, UMR-S 1172, 1 place de Verdun, F-59000 Lille, France
- University of Lille, University Hospital Federations (FHU) 1,000 days for Health, School of Medicine, 1 place de Verdun, F-59000 Lille, France
| | - John Garthwaite
- The Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Vincent Prevot
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, UMR-S 1172, 1 place de Verdun, F-59000 Lille, France
- University of Lille, University Hospital Federations (FHU) 1,000 days for Health, School of Medicine, 1 place de Verdun, F-59000 Lille, France
| |
Collapse
|
21
|
Wu MV, Tollkuhn J. Estrogen receptor alpha is required in GABAergic, but not glutamatergic, neurons to masculinize behavior. Horm Behav 2017; 95:3-12. [PMID: 28734725 PMCID: PMC7011612 DOI: 10.1016/j.yhbeh.2017.07.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 06/05/2017] [Accepted: 07/04/2017] [Indexed: 01/06/2023]
Abstract
Masculinization of the altricial rodent brain is driven by estrogen signaling during a perinatal critical period. Genetic deletion of estrogen receptor alpha (Esr1/ERα) results in altered hypothalamic-pituitary-gonadal (HPG) axis signaling and a dramatic reduction of male sexual and territorial behaviors. However, the role of ERα in masculinizing distinct classes of neurons remains unexplored. We deleted ERα in excitatory or inhibitory neurons using either a Vglut2 or Vgat driver and assessed male behaviors. We find that Vglut2-Cre;Esr1lox/lox mutant males lack ERα in the ventrolateral region of the ventromedial hypothalamus (VMHvl) and posterior ventral portion of the medial amygdala (MePV). These mutants recapitulate the increased serum testosterone levels seen with constitutive ERα deletion, but have none of the behavioral deficits. In contrast, Vgat-Cre;Esr1lox/lox males with substantial ERα deletion in inhibitory neurons, including those of the principal nucleus of the bed nucleus of the stria terminalis (BNSTpr), posterior dorsal MeA (MePD), and medial preoptic area (MPOA) have normal testosterone levels, but display alterations in mating and territorial behaviors. These mutants also show dysmasculinized expression of androgen receptor (AR) and estrogen receptor beta (Esr2). Our results demonstrate that ERα masculinizes GABAergic neurons that gate the display of male-typical behaviors.
Collapse
Affiliation(s)
- Melody V Wu
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Jessica Tollkuhn
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
22
|
Grassi D, Lagunas N, Pinos H, Panzica G, Garcia-Segura LM, Collado P. NADPH-Diaphorase Colocalizes with GPER and Is Modulated by the GPER Agonist G1 in the Supraoptic and Paraventricular Nuclei of Ovariectomized Female Rats. Neuroendocrinology 2017; 104:94-104. [PMID: 26954778 DOI: 10.1159/000445190] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 03/02/2016] [Indexed: 11/19/2022]
Abstract
Nitric oxide is produced in the brain by the neuronal nitric oxide synthase (nNOS) and carries out a wide range of functions by acting as a neurotransmitter-like molecule. Gonadal hormones are involved in the regulation of the brain nitrergic system. We have previously demonstrated that estradiol, via classical estrogen receptors (ERs), regulates NOS activity in the supraoptic (SON) and paraventricular (PVN) nuclei of the hypothalamus, acting through both ERα and ERβ. Magnocellular and parvocellular neurons in the SON and PVN also express the G protein-coupled ER (GPER). In this study, we have assessed whether GPER is also involved in the regulation of nicotinamide adenine dinucleotide phosphate (NADPH)-diaphorase in the SON and PVN. Adult female ovariectomized rats were treated with G1, a selective GPER agonist, or with G1 in combination with G15, a selective GPER antagonist. G1 treatment decreased NADPH-diaphorase expression in the SON and in all PVN subnuclei. The treatment with G1 + G15 effectively rescued the G1-dependent decrease in NADPH-diaphorase expression in both brain regions. In addition, the activation of extracellular signal-regulated kinase (ERK) 1/2, one of the kinases involved in the GPER-dependent intracellular signaling pathway and in NOS phosphorylation, was assessed in the same brain nuclei. Treatment with G1 significantly decreased the number of p-ERK 1/2-positive cells in the SON and PVN, while the treatment with G1 + G15 significantly recovered its number to control values. These findings suggest that the activation of GPER in the SON and PVN inhibits the phosphorylation of ERK 1/2, which induces a decrease in NADPH-diaphorase expression.
Collapse
Affiliation(s)
- Daniela Grassi
- Department of Psychobiology, Universidad Nacional de Educacion a Distancia (UNED), Madrid, Spain
| | | | | | | | | | | |
Collapse
|
23
|
Goldstein JM. Prenatal stress-immune programming of sex differences in comorbidity of depression and obesity/metabolic syndrome. DIALOGUES IN CLINICAL NEUROSCIENCE 2016; 18:425-436. [PMID: 28179814 PMCID: PMC5286728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2023]
Abstract
Major depressive disorder (MDD) is the number one cause of disability worldwide and is comorbid with many chronic diseases, including obesity/metabolic syndrome (MetS). Women have twice as much risk for MDD and comorbidity with obesity/MetS as men, although pathways for understanding this association remain unclear. On the basis of clinical and preclinical studies, we argue that prenatal maternal stress (ie, excess glucocorticoid expression and associated immune responses) that occurs during the sexual differentiation of the fetal brain has sex-dependent effects on brain development within highly sexually dimorphic regions that regulate mood, stress, metabolic function, the autonomic nervous system, and the vasculature. Furthermore, these effects have lifelong consequences for shared sex-dependent risk of MDD and obesity/MetS. Thus, we propose that there are shared biologic substrates at the anatomical, molecular, and/or genetic levels that produce the comorbid risk for MDD-MetS through sex-dependent fetal origins.
Collapse
Affiliation(s)
- Jill M. Goldstein
- Connors Center for Women's Health and Gender Biology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Departments of Psychiatry and Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Huilgol D, Tole S. Cell migration in the developing rodent olfactory system. Cell Mol Life Sci 2016; 73:2467-90. [PMID: 26994098 PMCID: PMC4894936 DOI: 10.1007/s00018-016-2172-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 02/08/2016] [Accepted: 03/01/2016] [Indexed: 02/06/2023]
Abstract
The components of the nervous system are assembled in development by the process of cell migration. Although the principles of cell migration are conserved throughout the brain, different subsystems may predominantly utilize specific migratory mechanisms, or may display unusual features during migration. Examining these subsystems offers not only the potential for insights into the development of the system, but may also help in understanding disorders arising from aberrant cell migration. The olfactory system is an ancient sensory circuit that is essential for the survival and reproduction of a species. The organization of this circuit displays many evolutionarily conserved features in vertebrates, including molecular mechanisms and complex migratory pathways. In this review, we describe the elaborate migrations that populate each component of the olfactory system in rodents and compare them with those described in the well-studied neocortex. Understanding how the components of the olfactory system are assembled will not only shed light on the etiology of olfactory and sexual disorders, but will also offer insights into how conserved migratory mechanisms may have shaped the evolution of the brain.
Collapse
Affiliation(s)
- Dhananjay Huilgol
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
- Cold Spring Harbor Laboratory, Cold Spring Harbor, USA
| | - Shubha Tole
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India.
| |
Collapse
|
25
|
Govindaraj V, Rao AJ. Proteomic identification of non-erythrocytic alpha-spectrin-1 down-regulation in the pre-optic area of neonatally estradiol-17β treated female adult rats. Horm Mol Biol Clin Investig 2016; 26:165-72. [PMID: 27166725 DOI: 10.1515/hmbci-2016-0008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/21/2016] [Indexed: 11/15/2022]
Abstract
It is well established that sexually dimorphic brain regions, which are critical for reproductive physiology and behavior, are organized by steroid hormones during the first 2 weeks after birth in the rodents. In our recent observation, neonatal exposure to estradiol-17β (E2) in the female rat revealed increase in cyclooxygenase 2 (COX-2) level, sexually dimorphic nucleus (SDN)-pre-optic area (POA) size and down-regulation of synaptogenesis related genes in POA in the adult stage. In the present study, using the same animal model, the protein profile of control and neonatally E2-treated POA was compared by 1D-SDS-PAGE, and the protein that shows a change in abundance was identified by LC-MS/MS analysis. Results indicated that there was a single protein band, which was down-regulation in E2-treated POA and it was identified as spectrin alpha chain, non-erythrocytic 1 (SPTAN1). Consistently, the down-regulation of SPTAN1 expression was also confirmed by reverse transcription polymerase chain reaction (RT-PCR) and Western blot analysis. The SPTAN1 was identified as a cytoskeletal protein that is involved in stabilization of the plasma membrane and organizes intracellular organelles, and it has been implicated in cellular functions including DNA repair and cell cycle regulation. The evidence shows that any mutation in spectrins causes impairment of synaptogenesis and other neurological disorders. Also, protein-protein interaction analysis of SPTAN1 revealed a strong association with proteins such as kirrel, actinin, alpha 4 (ACTN4) and vinculin (VCL) which are implicated in sexual behavior, masculinization and defeminization. Our results indicate that SPTAN1 expression in the developing rat brain is sexually dimorphic, and we suggest that this gene may mediate E2-17β-induced masculinization and defeminization, and disrupted reproductive function in the adult stage.
Collapse
|
26
|
Senthilkumaran B, Sudhakumari CC, Mamta SK, Raghuveer K, Swapna I, Murugananthkumar R. "Brain sex differentiation" in teleosts: Emerging concepts with potential biomarkers. Gen Comp Endocrinol 2015; 220:33-40. [PMID: 26116093 DOI: 10.1016/j.ygcen.2015.06.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 06/05/2015] [Accepted: 06/08/2015] [Indexed: 12/13/2022]
Abstract
"Brain sex differentiation" in teleosts is a contentious topic of research as most of the earlier reports tend to suggest that gonadal sex differentiation drives brain sex differentiation. However, identification of sex-specific marker genes in the developing brain of teleosts signifies brain-gonadal interaction during early sexual development in lower vertebrates. In this context, the influence of gonadotropin-releasing hormone (GnRH)-gonadotropin (GTH) axis on gonadal sex differentiation, if any requires in depth analysis. Presence of seabream (sb) GnRH immunoreactivity (ir-) in the brain of XY Nile tilapia was found as early as 5days post hatch (dph) followed by qualitative reduction in the preoptic area-hypothalamus region. In contrast, in the XX female brain a steady ir- of sbGnRH was evident from 15dph. Earlier studies using sea bass already implied the importance of hypothalamic gonadotropic axis completion during sex differentiation period. Such biphasic pattern of localization was also seen in pituitary GTHs using heterologous antisera in tilapia. However, more recent analysis in the same species could not detect any sexually dimorphic pattern using homologous antisera for pituitary GTHs. Detailed studies on the development of hypothalamo-hypophyseal-gonadal axis in teleosts focusing on hypothalamic monoamines (MA) and MA-related enzymes demonstrated sex-specific differential expression of tryptophan hydroxylase (Tph) in the early stages of developing male and female brains of tilapia and catfish. The changes in Tph expression was in agreement with the levels of serotonin (5-HT) and 5-hydroxytryptophan in the preoptic area-hypothalamus. Considering the stimulatory influence of 5-HT on GnRH and GTH release, it is possible to propose a network association between these correlates during early development, which may bring about brain sex dimorphism in males. A recent study from our laboratory during female brain sex development demonstrated high expression of tyrosine hydroxylase in correlation with catecholamine levels, brain aromatase and its related transcription factors such as fushi tarazu factor 1, Ftz-f1 and fork head box protein L2, foxl2. Taken together, gender differences in the levels of various transcripts provide new perspectives on brain sex differentiation in lower vertebrates. Sexually dimorphic or differentially expressing genes may play an essential role at the level of brain in response to gonadal differentiation, which might consequentially or causatively respond to gonadal sex.
Collapse
Affiliation(s)
- Balasubramanian Senthilkumaran
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, P.O. Central University, Hyderabad 500046, Telangana, India.
| | - Cheni-Chery Sudhakumari
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, P.O. Central University, Hyderabad 500046, Telangana, India
| | - Sajwan-Khatri Mamta
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, P.O. Central University, Hyderabad 500046, Telangana, India
| | - Kavarthapu Raghuveer
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, P.O. Central University, Hyderabad 500046, Telangana, India
| | - Immani Swapna
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, P.O. Central University, Hyderabad 500046, Telangana, India
| | - Raju Murugananthkumar
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, P.O. Central University, Hyderabad 500046, Telangana, India
| |
Collapse
|
27
|
Case SS, Oetama-Paul AJ. Brain Biology and Gendered Discourse. APPLIED PSYCHOLOGY-AN INTERNATIONAL REVIEW-PSYCHOLOGIE APPLIQUEE-REVUE INTERNATIONALE 2014. [DOI: 10.1111/apps.12040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Susan S. Case
- Weatherhead School of Management; Case Western Reserve University; USA
| | | |
Collapse
|
28
|
Goldstein JM, Holsen L, Handa R, Tobet S. Fetal hormonal programming of sex differences in depression: linking women's mental health with sex differences in the brain across the lifespan. Front Neurosci 2014; 8:247. [PMID: 25249929 PMCID: PMC4157606 DOI: 10.3389/fnins.2014.00247] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/24/2014] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jill M Goldstein
- Division of Women's Health, Departments of Psychiatry and Medicine, Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital Boston, MA, USA ; Departments of Psychiatry and Medicine, Harvard Medical School Boston, MA, USA ; Division of Psychiatric Neuroscience, Department of Psychiatry, Massachusetts General Hospital Boston, MA, USA
| | - Laura Holsen
- Division of Women's Health, Departments of Psychiatry and Medicine, Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital Boston, MA, USA ; Departments of Psychiatry and Medicine, Harvard Medical School Boston, MA, USA
| | - Robert Handa
- Department of Basic Medical Sciences, University of Arizona College of Medicine Phoenix, AZ, USA
| | - Stuart Tobet
- Department of Biomedical Sciences and School of Biomedical Engineering, College of Veterinary Medicine and Biomedical Sciences, Colorado State University Fort Collins, CO, USA
| |
Collapse
|
29
|
Reddy RC, Scheldrup M, Meaker M, Stormshak F, Estill CT, Roselli CE. Cell death in the central division of the medial preoptic nucleus of male and female lamb fetuses. Brain Res 2014; 1554:21-8. [PMID: 24491631 DOI: 10.1016/j.brainres.2014.01.041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 01/22/2014] [Accepted: 01/25/2014] [Indexed: 10/25/2022]
Abstract
The medial preoptic area of the adult sheep contains an ovine sexually dimorphic nucleus (oSDN) that is larger and has more neurons in males than in females. In the lamb fetus, the nascent oSDN occupies the central division of the medial preoptic nucleus (MPNc) and consists of a cluster of cells that is organized by the action of testosterone during gestational days 60-90 of a 147 day term pregnancy. The current study sought to determine whether programmed cell death contributes to the emergence of the oSDN. Male and female lamb fetuses were euthanized at different ages spanning the period during which the oSDN is organized. The expression of the pro- and anti-apoptotic genes bcl-2 and bax, respectively, was measured by quantitative RT-PCR to assess possible sex differences in neuron vulnerability to programmed cell death. The appearance of DNA-fragmentation was detected by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and used to estimate the occurrence of apoptotic cell death. We found that bcl-2 and bax mRNA expression in the medial preoptic area of the developing lamb fetus decreased during the last half of the 147-day gestation. The ratio of bcl-2/bax gene expression was highest at gestational day 85 but was equivalent between males and females. TUNEL staining in the MPNc was very low and although it decreased significantly with age, it was not significantly different between sexes. These results using two different methods to assess cell death indicate that a sex difference in the incidence of cell death is not a primary mechanism leading to sexual differentiation of the oSDN.
Collapse
Affiliation(s)
- Radhika C Reddy
- Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Melissa Scheldrup
- Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Mary Meaker
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Fred Stormshak
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Charles T Estill
- College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Charles E Roselli
- Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA.
| |
Collapse
|
30
|
Gender identity disorder and schizophrenia: neurodevelopmental disorders with common causal mechanisms? SCHIZOPHRENIA RESEARCH AND TREATMENT 2014; 2014:463757. [PMID: 25548672 PMCID: PMC4274821 DOI: 10.1155/2014/463757] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/20/2014] [Indexed: 12/15/2022]
Abstract
Gender identity disorder (GID), recently renamed gender dysphoria (GD), is a rare condition characterized by an incongruity between gender identity and biological sex. Clinical evidence suggests that schizophrenia occurs in patients with GID at rates higher than in the general population and that patients with GID may have schizophrenia-like personality traits. Conversely, patients with schizophrenia may experience alterations in gender identity and gender role perception. Neurobiological research, including brain imaging and studies of finger length ratio and handedness, suggests that both these disorders are associated with altered cerebral sexual dimorphism and changes in cerebral lateralization. Various mechanisms, such as Toxoplasma infection, reduced levels of brain-derived neurotrophic factor (BDNF), early childhood adversity, and links with autism spectrum disorders, may account for some of this overlap. The implications of this association for further research are discussed.
Collapse
|
31
|
de Vries GJ, Fields CT, Peters NV, Whylings J, Paul MJ. Sensitive periods for hormonal programming of the brain. Curr Top Behav Neurosci 2014; 16:79-108. [PMID: 24549723 DOI: 10.1007/7854_2014_286] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During sensitive periods, information from the external and internal environment that occurs during particular phases of development is relayed to the brain to program neural development. Hormones play a central role in this process. In this review, we first discuss sexual differentiation of the brain as an example of hormonal programming. Using sexual differentiation, we define sensitive periods, review cellular and molecular processes that can explain their restricted temporal window, and discuss challenges in determining the precise timing of the temporal window. We then briefly review programming effects of other hormonal systems and discuss how programming of these systems interact with sexual differentiation.
Collapse
Affiliation(s)
- Geert J de Vries
- Neuroscience Institute, Georgia State University, PO Box 5030, Atlanta, GA, 30302-5030, USA,
| | | | | | | | | |
Collapse
|
32
|
Goldstein JM, Handa RJ, Tobet SA. Disruption of fetal hormonal programming (prenatal stress) implicates shared risk for sex differences in depression and cardiovascular disease. Front Neuroendocrinol 2014; 35:140-58. [PMID: 24355523 PMCID: PMC3917309 DOI: 10.1016/j.yfrne.2013.12.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 10/31/2013] [Accepted: 12/04/2013] [Indexed: 12/19/2022]
Abstract
Comorbidity of major depressive disorder (MDD) and cardiovascular disease (CVD) represents the fourth leading cause of morbidity and mortality worldwide, and women have a two times greater risk than men. Thus understanding the pathophysiology has widespread implications for attenuation and prevention of disease burden. We suggest that sex-dependent MDD-CVD comorbidity may result from alterations in fetal programming consequent to the prenatal maternal environments that produce excess glucocorticoids, which then drive sex-dependent developmental alterations of the fetal hypothalamic-pituitary-adrenal (HPA) axis circuitry impacting mood, stress regulation, autonomic nervous system (ANS), and the vasculature in adulthood. Evidence is consistent with the hypothesis that disruptions of pathways associated with gamma aminobutyric acid (GABA) in neuronal and vascular development and growth factors have critical roles in key developmental periods and adult responses to injury in heart and brain. Understanding the potential fetal origins of these sex differences will contribute to development of novel sex-dependent therapeutics.
Collapse
Affiliation(s)
- J M Goldstein
- Departments of Psychiatry and Medicine, Harvard Medical School, Boston, MA, USA; Brigham and Women's Hospital (BWH), Connors Center for Women's Health & Gender Biology, 1620 Tremont St. BC-3-34, Boston, MA 02120, USA; BWH, Departments of Psychiatry and Medicine, 1620 Tremont St. BC-3-34, Boston, MA 02120, USA.
| | - R J Handa
- Department of Basic Medical Sciences, University of Arizona College of Medicine, 425 N. Fifth Street, Phoenix, AZ 85004, USA
| | - S A Tobet
- Department of Biomedical Sciences and School of Biomedical Engineering, Colorado State University, 1617 Campus Delivery, Fort Collins, CO 80523, USA
| |
Collapse
|
33
|
Messina A, Giacobini P. Semaphorin signaling in the development and function of the gonadotropin hormone-releasing hormone system. Front Endocrinol (Lausanne) 2013; 4:133. [PMID: 24065959 PMCID: PMC3779810 DOI: 10.3389/fendo.2013.00133] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 09/09/2013] [Indexed: 12/17/2022] Open
Abstract
The semaphorin proteins are among the best-studied families of guidance cues, contributing to morphogenesis and homeostasis in a wide range of tissue types. The major semaphorin receptors are plexins and neuropilins, however other receptors and co-receptors are capable to mediate signaling by semaphorins. These guidance proteins were originally identified as growth cone "collapsing factors" or as inhibitory signals, crucial for nervous system development. Since those seminal discoveries, the list of functions of semaphorins has rapidly grown. Over the past few years, a growing body of data indicates that semaphorins are involved in the regulation of the immune and vascular systems, in tumor growth/cancer cell metastasis and in neural circuit formation. Recently there has been increasing emphasis on research to determine the potential influence of semaphorins on the development and homeostasis of hormone systems and how circulating reproductive hormones regulate their expression and functions. Here, we focus on the emerging role of semaphorins in the development, differentiation and plasticity of unique neurons that secrete gonadotropin-releasing hormone (GnRH), which are essential for the acquisition and maintenance of reproductive competence in all vertebrates. Genetic evidence is also provided showing that insufficient semaphorin signaling contributes to some forms of reproductive disorders in humans, characterized by the reduction or failure of sexual competence. Finally, we will review some studies with the goal of highlighting how the expression of semaphorins and their receptors might be regulated by gonadal hormones in physiological and pathological conditions.
Collapse
Affiliation(s)
- Andrea Messina
- INSERM, Laboratory of Development and Plasticity of the Postnatal Brain, Jean-Pierre Aubert Research Center, Unité 837, Lille, France
- School of Medicine, UDSL, Lille, France
| | - Paolo Giacobini
- INSERM, Laboratory of Development and Plasticity of the Postnatal Brain, Jean-Pierre Aubert Research Center, Unité 837, Lille, France
- School of Medicine, UDSL, Lille, France
- *Correspondence: Paolo Giacobini, INSERM, Laboratory of Development and Plasticity of the Postnatal Brain, Jean-Pierre Aubert Research Center, Unit 837, Place de Verdun, 59045 Lille Cedex, France e-mail:
| |
Collapse
|
34
|
Patra PB, Patra S. Sex differences in the physiology and pharmacology of the lower urinary tract. Curr Urol 2013; 6:179-88. [PMID: 24917740 DOI: 10.1159/000343536] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Accepted: 12/04/2012] [Indexed: 12/13/2022] Open
Abstract
Sexual dimorphism is not only noticed in the prevalence of many diseases, but also in multiple physiological functions in the body. This review has summarized findings from published literature on the sex differences of the pathophysiology and pharmacology of the lower urinary tract (LUT) of humans and animals. Sex differences have been found in several key areas of the LUT, such as overactive bladder, expression and function of neurotransmitter receptors in the bladder and urethra, and micturition patterns in humans and animals. It is anticipated that this review will not only evoke renewed interest for further research on the mechanism of sex differences in the pathophysiology of the LUT (especially for overactive bladder), but might also open up the possibilities for gender-based drug development by pharmaceutical industries in order to find separate cures for men and women with diseases of the LUT.
Collapse
|
35
|
Suppression of β1-integrin in gonadotropin-releasing hormone cells disrupts migration and axonal extension resulting in severe reproductive alterations. J Neurosci 2013; 32:16992-7002. [PMID: 23175850 DOI: 10.1523/jneurosci.3057-12.2012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Reproduction in mammals is dependent on the function of hypothalamic neurons whose axons project to the hypothalamic median eminence (ME) where they release gonadotropin-releasing hormone (GnRH) into a specialized capillary network for delivery to the anterior pituitary. These neurons originate prenatally in the nasal placode and migrate into the forebrain along the olfactory-vomeronasal nerves. The complex developmental events leading to the correct establishment of the GnRH system are tightly regulated by the specific spatiotemporal expression patterns of guidance cues and extracellular matrix molecules, the functions of which, in part, are mediated by their binding to β1-subunit-containing integrins. To determine the biological role of these cell-surface proteins in reproduction, Cre/LoxP technology was used to generate GnRH neuron-specific β1-integrin conditional KO (GnRH-Itgb1(-/-)) mice. Loss of β1-integrin signaling impaired migration of GnRH neurons, their axonal extension to the ME, timing of pubertal onset, and fertility in these mice. These results identify β1-integrin as a gene involved in normal development of the GnRH system and demonstrate a fundamental role for this protein in acquisition of normal reproductive competence in female mice.
Collapse
|
36
|
Volumetric parcellation methodology of the human hypothalamus in neuroimaging: normative data and sex differences. Neuroimage 2012; 69:1-10. [PMID: 23247186 DOI: 10.1016/j.neuroimage.2012.12.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 11/30/2012] [Accepted: 12/01/2012] [Indexed: 11/22/2022] Open
Abstract
There is increasing evidence regarding the importance of the hypothalamus for understanding sex differences in relation to neurological, psychiatric, endocrine and sleep disorders. Although different in histology, physiology, connections and function, multiple hypothalamic nuclei subserve non-voluntary functions and are nodal points for the purpose of maintaining homeostasis of the organism. Thus, given the critical importance of hypothalamic nuclei and their key multiple roles in regulating basic functions, it is important to develop the ability to conduct in vivo human studies of anatomic structure, volume, connectivity, and function of hypothalamic regions represented at the level of its nuclei. The goals of the present study were to develop a novel method of semi-automated volumetric parcellation for the human hypothalamus that could be used to investigate clinical conditions using MRI and to demonstrate its applicability. The proposed new method subdivides the hypothalamus into five parcels based on visible anatomic landmarks associated with specific nuclear groupings and was confirmed using two ex vivo hypothalami that were imaged in a 7 T (7 T) scanner and processed histologically. Imaging results were compared with histology from the same brain. Further, the method was applied to 44 healthy adults (26 men; 18 women, comparable on age, handedness, ethnicity, SES) to derive normative volumes and assess sex differences in hypothalamic regions using 1.5 T MRI. Men compared to women had a significantly larger total hypothalamus, relative to cerebrum size, similar for both hemispheres, a difference that was primarily driven by the tuberal region, with the sex effect size being largest in the superior tuberal region and, to a lesser extent, inferior tuberal region. Given the critical role of hypothalamic nuclei in multiple chronic diseases and the importance of sex differences, we argue that the use of the novel methodology presented here will allow for critical investigations of these disorders and further delineation of potential treatments, particularly sex-specific approaches to gene and drug discoveries that involve hypothalamic nuclei.
Collapse
|
37
|
Zhang Q, Bouma GJ, McClellan K, Tobet S. Hypothalamic expression of snoRNA Snord116 is consistent with a link to the hyperphagia and obesity symptoms of Prader-Willi syndrome. Int J Dev Neurosci 2012; 30:479-85. [PMID: 22664655 DOI: 10.1016/j.ijdevneu.2012.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 05/23/2012] [Accepted: 05/24/2012] [Indexed: 01/07/2023] Open
Abstract
The hypothalamus is integral to the regulation of body homeostasis, including food intake, energy balance, and blood pressure. Dysfunction of the hypothalamus has been associated with a broad range of disorders; many of which are sex-dependent in prevalence. Small nucleolar (sno) RNAs are a group of small RNAs located in nucleoli that modulate chemical modifications and maturation of ribosomal or other RNAs. Recent data suggest that snoRNA Snord116 is important for the pathogenesis of Prader-Willi syndrome (PWS) characterized by hyperphagia and obesity. The current study was conducted to assess a potential cellular link between Snord116 and phenotypes of PWS. Data from mice revealed Snord116 expression in the medial hypothalamus, particularly within nuclei that are part of feeding circuitry. High expression of Snord116 was evident in the paraventricular (PVN) and ventromedial (VMH) nuclei, but particularly prevalent in the arcuate nucleus (ARC) according to in situ hybridization. Snord116 expression level in ventral hypothalamic dissections including ARC was significantly greater (by 2-fold) than that in cortex; and its expression level in dorsal hypothalamic dissections including PVN was double that in cortex. The enhanced expression pattern of Snord116 in hypothalamic nuclei was observed at weaning and young adult stages, but less obvious neonatally when expression was significantly more widespread. Therefore the expression of Snord116 likely is regulated developmentally. These results provide a new lead for understanding the mechanism(s) related to hyperphagia and obesity symptoms in PWS patients.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | | | | | | |
Collapse
|
38
|
Lee TW, Yu YWY, Hong CJ, Tsai SJ, Wu HC, Chen TJ. The influence of dopamine receptor d4 polymorphism on resting EEG in healthy young females. Open Neuroimag J 2012; 6:19-25. [PMID: 22448208 PMCID: PMC3308261 DOI: 10.2174/1874440001206010019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 12/22/2011] [Accepted: 01/24/2012] [Indexed: 01/15/2023] Open
Abstract
The polymorphism of variable number of tandem repeat (VNTR) in dopamine receptor D4 (DRD4) gene exon III has been linked to various neuro-psychiatric conditions with disinhibition/impulsivity as one of the core features. This study examined the modulatory effects of long-allele variant of DRD4 VNTR on the regional neural activity as well as inter-regional neural interactions in a young female population. Blood sample and resting state eyes-closed EEG signals were collected in 233 healthy females, stratified into two groups by polymerase chain reaction: long-allele carriers (>4- repeat) and non-carriers (<=4-repeat/<=4-repeat). The values of mean power of 18 electrodes and mutual information of 38 channel pairs across theta, alpha, and beta frequencies were analyzed. Our connectivity analysis was based on information theory, which combined Morlet wavelet transform and mutual information calculation. Between-group differences of regional power and connectivity strength were quantified by independent t-test, while between-group differences in global trends were examined by non-parametric analyses. We noticed that DRD4 VNTR long-allele was associated with decreased global connectivity strength (from non-parametric analysis), especially over bi-frontal, biparietal and right fronto-parietal and right fronto-temporal connections (from independent t-tests). The between-group differences in regional power were not robust. Our findings fit with the networks of response inhibition, providing evidence bridging DRD4 long-allele and disinhibition/impulsivity in neuropsychiatric disorders. We suggest future DRD4 studies of imaging genetics incorporate connectivity analysis to unveil its impact on cerebral network.
Collapse
Affiliation(s)
- Tien-Wen Lee
- Laureate Institute for Brain Research, Tulsa, OK 74136, USA
| | | | | | | | | | | |
Collapse
|
39
|
Petersen SL, Krishnan S, Aggison LK, Intlekofer KA, Moura PJ. Sexual differentiation of the gonadotropin surge release mechanism: a new role for the canonical NfκB signaling pathway. Front Neuroendocrinol 2012; 33:36-44. [PMID: 21741397 DOI: 10.1016/j.yfrne.2011.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 06/09/2011] [Accepted: 06/11/2011] [Indexed: 12/30/2022]
Abstract
Sex differences in luteinizing hormone (LH) release patterns are controlled by the hypothalamus, established during the perinatal period and required for fertility. Female mammals exhibit a cyclic surge pattern of LH release, while males show a tonic release pattern. In rodents, the LH surge pattern is dictated by the anteroventral periventricular nucleus (AVPV), an estrogen receptor-rich structure that is larger and more cell-dense in females. Sex differences result from mitochondrial cell death triggered in perinatal males by estradiol derived from aromatization of testosterone. Herein we provide an historical perspective and an update describing evidence that molecules important for cell survival and cell death in the immune system also control these processes in the developing AVPV. We conclude with a new model proposing that development of the female AVPV requires constitutive activation of the Tnfα, Tnf receptor 2, NfκB and Bcl2 pathway that is blocked by induction of Tnf receptor-associated factor 2-inhibiting protein (Traip) in the male.
Collapse
Affiliation(s)
- Sandra L Petersen
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003, United States.
| | | | | | | | | |
Collapse
|
40
|
deCampo D, Fudge J. Where and what is the paralaminar nucleus? A review on a unique and frequently overlooked area of the primate amygdala. Neurosci Biobehav Rev 2012; 36:520-35. [PMID: 21906624 PMCID: PMC3221880 DOI: 10.1016/j.neubiorev.2011.08.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 07/25/2011] [Accepted: 08/18/2011] [Indexed: 12/16/2022]
Abstract
The primate amygdala is composed of multiple subnuclei that play distinct roles in amygdala function. While some nuclei have been areas of focused investigation, others remain virtually unknown. One of the more obscure regions of the amygdala is the paralaminar nucleus (PL). The PL in humans and non-human primates is relatively expanded compared to lower species. Long considered to be part of the basal nucleus, the PL has several interesting features that make it unique. These features include a dense concentration of small cells, high concentrations of receptors for corticotropin releasing hormone and benzodiazepines, and dense innervation of serotonergic fibers. More recently, high concentrations of immature-appearing cells have been noted in the primate PL, suggesting special mechanisms of neural plasticity. Following a brief overview of amygdala structure and function, this review will provide an introduction to the history, embryology, anatomical connectivity, immunohistochemical and cytoarchitectural properties of the PL. Our conclusion is that the PL is a unique subregion of the amygdala that may yield important clues about the normal growth and function of the amygdala, particularly in higher species.
Collapse
Affiliation(s)
| | - Julie Fudge
- Department of Neurobiology and Anatomy
- Department of Psychiatry
| |
Collapse
|
41
|
Aiello TP, Whitaker-Azmitia PM. Sexual differentiation and the neuroendocrine hypothesis of autism. Anat Rec (Hoboken) 2011; 294:1663-70. [PMID: 21901838 DOI: 10.1002/ar.21251] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 05/25/2010] [Indexed: 11/09/2022]
Abstract
The phenotypic expression of autism spectrum disorders varies widely in severity and characteristics and it is, therefore, likely that a number of etiological factors are involved. However, one finding which has been found consistently is that there is a greater incidence of autism in boys than girls. Recently, attention has been given to the extreme male hypothesis-that is that autism behaviors are an extreme form of typical male behaviors, including lack of empathy and language deficits but an increase in so-called systemizing behaviors, such as attention to detail and collecting. This points to the possibility that an alteration during sexual differentiation of the brain may occur in autism. During sexual differentiation of the brain, two brain regions are highly sexually dimorphic-the amygdala and the hypothalamus. Both of these regions are also implicated in the neuroendocrine hypothesis of autism, wherein a balance between oxytocin and cortisol may contribute to the disorder. We are thus proposing that the extreme male hypothesis and the neuroendocrine hypothesis are in fact compatible in that sexual differentiation of the brain towards an extreme male phenotype would result in the neuroendocrine changes proposed in autism. We have preliminary data, treating developing rat pups with the differentiating hormone 17-β estradiol during a critical time and showing changes in social behaviors and oxytocin, to support this hypothesis. Further studies should be undertaken to confirm the role of extremes of normal sexual differentiation in producing the neuroendocrine changes associated with autism.
Collapse
|
42
|
Numakawa T, Matsumoto T, Numakawa Y, Richards M, Yamawaki S, Kunugi H. Protective Action of Neurotrophic Factors and Estrogen against Oxidative Stress-Mediated Neurodegeneration. J Toxicol 2011; 2011:405194. [PMID: 21776259 PMCID: PMC3135156 DOI: 10.1155/2011/405194] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 02/28/2011] [Accepted: 03/29/2011] [Indexed: 01/01/2023] Open
Abstract
Oxidative stress is involved in the pathogenesis of neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. Low levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) are important for maintenance of neuronal function, though elevated levels lead to neuronal cell death. A complex series of events including excitotoxicity, Ca(2+) overload, and mitochondrial dysfunction contributes to oxidative stress-mediated neurodegeneration. As expected, many antioxidants like phytochemicals and vitamins are known to reduce oxidative toxicity. Additionally, growing evidence indicates that neurotrophic factors such as brain-derived neurotrophic factor (BDNF) and estrogens significantly prevent neuronal damage caused by oxidative stress. Here, we review and discuss recent studies addressing the protective mechanisms of neurotrophic factors and estrogen within this system.
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
| | - Tomoya Matsumoto
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
- Department of Psychiatry and Neurosciences, Division of Frontier Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Yumiko Numakawa
- Peptide-prima Co., Ltd., 1-25-81, Nuyamazu, Kumamoto 861-2102, Japan
| | - Misty Richards
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
- The Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, NY 12208, USA
| | - Shigeto Yamawaki
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
- Department of Psychiatry and Neurosciences, Division of Frontier Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
| |
Collapse
|
43
|
Brabant G, Cain J, Jackson A, Kreitschmann-Andermahr I. Visualizing hormone actions in the brain. Trends Endocrinol Metab 2011; 22:153-63. [PMID: 21497512 DOI: 10.1016/j.tem.2011.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 01/19/2011] [Accepted: 01/20/2011] [Indexed: 01/01/2023]
Abstract
Profound and multifaceted effects of hormones on the development, maturation and function of the CNS are well documented. Recent developments in magnetic resonance imagining (MRI) and positron emission tomography (PET) permit detailed in vivo studies of cerebral structure and function in humans. Techniques to measure subtle differences in cerebral structure, regional brain activation, changes in blood flow and other physiological biomarkers allow us to translate experimental evidence of hormone effects obtained from animal models to humans. Here we review the imaging techniques available to support studies of hormone effects on the CNS, emphasizing the recent developments of MRI. In summarizing the major current studies we discuss the potential of these techniques for an emerging new field in endocrinology.
Collapse
Affiliation(s)
- Georg Brabant
- Department of Endocrinology, The Christie, Manchester Academic Health Science Centre, Wilmslow Road, Manchester M20 4BX, UK.
| | | | | | | |
Collapse
|
44
|
Naninck EFG, Lucassen PJ, Bakker J. Sex differences in adolescent depression: do sex hormones determine vulnerability? J Neuroendocrinol 2011; 23:383-92. [PMID: 21418338 DOI: 10.1111/j.1365-2826.2011.02125.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Depression is one of the most common, costly and severe psychopathologies worldwide. Its incidence, however, differs significantly between the sexes, and depression rates in women are twice those of men. Interestingly, this sex difference emerges during adolescence. Although the adolescent period is characterised by major physical and behavioural transformations, it is unclear why the incidence of depression increases so dramatically in girls during this otherwise generally healthy developmental period. Although psychological and environmental factors are also involved, we discuss the neuroendocrinological factors determining adolescent vulnerability to depression. In particular, we address the role of sex steroids in mood regulation, hypothalamic-pituitary-adrenal axis maturation and sexual differentiation of the brain, with a focus on hippocampal plasticity.
Collapse
Affiliation(s)
- E F G Naninck
- Centre for Neuroscience, Swammerdam Institute of Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
45
|
Salu Y. The roots of sexual arousal and sexual orientation. Med Hypotheses 2011; 76:384-7. [DOI: 10.1016/j.mehy.2010.10.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 10/29/2010] [Indexed: 10/18/2022]
|
46
|
Krohmer RW, DeMarchi GA, Baleckaitis DD, Lutterschmidt DI, Mason RT. Brain nuclei in actively courting red-sided garter snakes: A paradigm of neural trimorphism. Physiol Behav 2011; 102:532-7. [DOI: 10.1016/j.physbeh.2010.12.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 12/15/2010] [Accepted: 12/21/2010] [Indexed: 12/17/2022]
|
47
|
Sex differences in synaptic plasticity in stress-responsive brain regions following chronic variable stress. Physiol Behav 2011; 104:242-7. [PMID: 21315096 DOI: 10.1016/j.physbeh.2011.01.024] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 01/10/2011] [Accepted: 01/27/2011] [Indexed: 11/23/2022]
Abstract
Increased stress responsiveness is implicated in the etiology of mood and anxiety disorders, including depression and post-traumatic stress disorder. Additionally, stress-related affective disorders have a higher incidence in women than men. Chronic stress in rodents produces numerous neuromorphological changes in a variety of limbic brain regions. Here, we examined the sex-dependent differences in presynaptic innervation of the paraventricular nucleus of the hypothalamus (PVN), prefrontal cortex (PFC), bed nucleus of the stria terminalis (BST), and amygdala in response to chronic variable stress (CVS). Following 14 days of CVS, the presynaptic protein synaptophysin was assessed in male and female rats. Our results demonstrate that synaptophysin staining density was higher in females than males in all brain areas evaluated, indicating sex differences in the organization of presynaptic innervation. After CVS, the PVN, principal nucleus of the BST (BSTpr), and basolateral nucleus of the amygdala (BLA) displayed significantly reduced synaptophysin density in females but not males. Furthermore, males showed an increase in synaptophysin in the PVN after CVS, suggesting a sex difference in the modulation of presynaptic inputs to the PVN following chronic stress. Overall, these data suggest marked sex differences in PVN, BSTpr, and BLA presynaptic innervation as a consequence of chronic stress, which may be associated with differential stress responsivity and perhaps susceptibility to pathologies in males and females.
Collapse
|
48
|
Prenatal PCBs disrupt early neuroendocrine development of the rat hypothalamus. Toxicol Appl Pharmacol 2011; 252:36-46. [PMID: 21277884 DOI: 10.1016/j.taap.2011.01.012] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 01/13/2011] [Accepted: 01/19/2011] [Indexed: 11/22/2022]
Abstract
Neonatal exposure to endocrine disrupting chemicals (EDCs) such as polychlorinated biphenyls (PCBs) can interfere with hormone-sensitive developmental processes, including brain sexual differentiation. We hypothesized that disruption of these processes by gestational PCB exposure would be detectable as early as the day after birth (postnatal day (P) 1) through alterations in hypothalamic gene and protein expression. Pregnant Sprague-Dawley rats were injected twice, once each on gestational days 16 and 18, with one of the following: DMSO vehicle; the industrial PCB mixture Aroclor 1221 (A1221); a reconstituted mixture of the three most prevalent congeners found in humans, PCB138, PCB153, and PCB180; or estradiol benzoate (EB). On P1, litter composition, anogenital distance (AGD), and body weight were assessed. Pups were euthanized for immunohistochemistry of estrogen receptor α (ERα) or TUNEL labeling of apoptotic cells or quantitative PCR of 48 selected genes in the preoptic area (POA). We found that treatment with EB or A1221 had a sex-specific effect on developmental apoptosis in the neonatal anteroventral periventricular nucleus (AVPV), a sexually dimorphic hypothalamic region involved in the regulation of reproductive neuroendocrine function. In this region, exposed females had increased numbers of apoptotic nuclei, whereas there was no effect of treatment in males. For ERα, EB treatment increased immunoreactive cell numbers and density in the medial preoptic nucleus (MPN) of both males and females, while A1221 and the PCB mixture had no effect. PCR analysis of gene expression in the POA identified nine genes that were significantly altered by prenatal EDC exposure, in a manner that varied by sex and treatment. These genes included brain-derived neurotrophic factor, GABA(B) receptors-1 and -2, IGF-1, kisspeptin receptor, NMDA receptor subunits NR2b and NR2c, prodynorphin, and TGFα. Collectively, these results suggest that the disrupted sexual differentiation of the POA by prenatal EDC exposures is already evident as early as the day after birth, effects that may change the trajectory of postnatal development and compromise adult reproductive function.
Collapse
|
49
|
Bale TL, Baram TZ, Brown AS, Goldstein JM, Insel TR, McCarthy MM, Nemeroff CB, Reyes TM, Simerly RB, Susser ES, Nestler EJ. Early life programming and neurodevelopmental disorders. Biol Psychiatry 2010; 68:314-9. [PMID: 20674602 PMCID: PMC3168778 DOI: 10.1016/j.biopsych.2010.05.028] [Citation(s) in RCA: 643] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 05/17/2010] [Accepted: 05/20/2010] [Indexed: 02/06/2023]
Abstract
For more than a century, clinical investigators have focused on early life as a source of adult psychopathology. Early theories about psychic conflict and toxic parenting have been replaced by more recent formulations of complex interactions of genes and environment. Although the hypothesized mechanisms have evolved, a central notion remains: early life is a period of unique sensitivity during which experience confers enduring effects. The mechanisms for these effects remain almost as much a mystery today as they were a century ago. Recent studies suggest that maternal diet can program offspring growth and metabolic pathways, altering lifelong susceptibility to diabetes and obesity. If maternal psychosocial experience has similar programming effects on the developing offspring, one might expect a comparable contribution to neurodevelopmental disorders, including affective disorders, schizophrenia, autism, and eating disorders. Due to their early onset, prevalence, and chronicity, some of these disorders, such as depression and schizophrenia, are among the highest causes of disability worldwide according to the World Health Organization 2002 report. Consideration of the early life programming and transcriptional regulation in adult exposures supports a critical need to understand epigenetic mechanisms as a critical determinant in disease predisposition. Incorporating the latest insight gained from clinical and epidemiological studies with potential epigenetic mechanisms from basic research, the following review summarizes findings from a workshop on Early Life Programming and Neurodevelopmental Disorders held at the University of Pennsylvania in 2009.
Collapse
|
50
|
McClellan KM, Stratton MS, Tobet SA. Roles for gamma-aminobutyric acid in the development of the paraventricular nucleus of the hypothalamus. J Comp Neurol 2010; 518:2710-28. [PMID: 20506472 PMCID: PMC2879086 DOI: 10.1002/cne.22360] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The development of the hypothalamic paraventricular nucleus (PVN) involves several factors that work together to establish a cell group that regulates neuroendocrine functions and behaviors. Several molecular markers were noted within the developing PVN, including estrogen receptors (ER), neuronal nitric oxide synthase (nNOS), and brain-derived neurotrophic factor (BDNF). By contrast, immunoreactive gamma-aminobutyric acid (GABA) was found in cells and fibers surrounding the PVN. Two animal models were used to test the hypothesis that GABA works through GABA(A) and GABA(B) receptors to influence the development of the PVN. Treatment with bicuculline to decrease GABA(A) receptor signaling from embryonic day (E) 10 to E17 resulted in fewer cells containing immunoreactive (ir) ERalpha in the region of the PVN vs. control. GABA(B)R1 receptor subunit knockout mice were used to examine the PVN at P0 without GABA(B) signaling. In female but not male GABA(B)R1 subunit knockout mice, the positions of cells containing ir ERalpha shifted from medial to lateral compared with wild-type controls, whereas the total number of ir ERalpha-containing cells was unchanged. In E17 knockout mice, ir nNOS cells and fibers were spread over a greater area. There was also a significant decrease in ir BDNF in the knockout mice in a region-dependent manner. Changes in cell position and protein expression subsequent to disruption of GABA signaling may be due, in part, to changes in nNOS and BDNF signaling. Based on the current study, the PVN can be added as another site where GABA exerts morphogenetic actions in development.
Collapse
Affiliation(s)
- Kristy M. McClellan
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523
- School of Science, Buena Vista University, Storm Lake, IA 50588
| | - Matthew S. Stratton
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Stuart A. Tobet
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523
| |
Collapse
|