1
|
Prado RCR, Oliveira TN, Saunders B, Foster R, Bella ZIKDJD, Kilpatrick MW, Asano RY, Hackney AC, Takito MY. Effects of the Menstrual Cycle Phase on Cortisol Responses to Maximum Exercise in Women With and Without Premenstrual Syndrome. ENDOCRINES 2025; 6:14. [PMID: 40190449 PMCID: PMC11970578 DOI: 10.3390/endocrines6010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
Objectives This study investigated the effects of the menstrual cycle phases on cortisol levels before and after a maximal incremental exercise test in women with and without premenstrual syndrome (PMS). Methods Nineteen healthy, active and eumenorrheic women completed five maximal incremental exercise tests; three of those were performed at specific phases of the menstrual cycle (i.e., menses, follicular, and luteal). The participants were allocated into two groups according to the presence of PMS (n = 11) or absence of PMS (NO-PMS, n = 8). Samples of blood were collected before and after each experimental test. A three-way ANOVA was conducted to compare the differences between menstrual cycle phases (i.e., menses, follicular, and luteal), time (before and after) and groups (PMS and NO-PMS). Results The results demonstrated an alteration of cortisol across the menstrual cycle, with cortisol levels significantly (p < 0.05) higher during the follicular phase (mean = 11.0 μg/dL, CI95% = 9.1, 12.9) compared to the luteal phase (mean = 8.6 μg/dL, CI95% = 7.2, 10.4) in the PMS and NO-PMS groups. There was no difference (p > 0.05) in cortisol levels for groups or time. Conclusions This study observed significant cortisol fluctuations across the menstrual cycle phases in women with and without PMS. Future studies should consider alternative maximal incremental test protocols and incorporate a more comprehensive hormonal profile to provide a deeper physiological understanding of this population.
Collapse
Affiliation(s)
- Raul Cosme Ramos Prado
- Department of Pedagogy of Human Movement, School of Physical Education and Sport, University of São Paulo, São Paulo 05508-030, Brazil
| | - Tamires Nunes Oliveira
- Applied Physiology and Nutrition Research Group—School of Physical Education and Sport, FMUSP, Universidade de São Paulo, São Paulo 05508-030, Brazil
- Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Bryan Saunders
- Applied Physiology and Nutrition Research Group—School of Physical Education and Sport, FMUSP, Universidade de São Paulo, São Paulo 05508-030, Brazil
- Center of Lifestyle Medicine, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil
- Nutrology Academy, Rio de Janeiro 22421-030, Brazil
| | - Roberta Foster
- Department of Gynecology, Federal University of São Paulo, São Paulo 04021-001, Brazil
| | | | | | - Ricardo Yukio Asano
- Department of Pedagogy of Human Movement, School of Physical Education and Sport, University of São Paulo, São Paulo 05508-030, Brazil
| | - Anthony C. Hackney
- Department of Exercise & Sport Science, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Monica Yuri Takito
- Department of Pedagogy of Human Movement, School of Physical Education and Sport, University of São Paulo, São Paulo 05508-030, Brazil
| |
Collapse
|
2
|
Du R, Liang T, Lu G. Modulation of empathic abilities by the interplay between estrogen receptors and arginine vasopressin. Neurosci Res 2025; 210:11-18. [PMID: 39245211 DOI: 10.1016/j.neures.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/05/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
This review examines the complex interactions between estrogen receptors α and β (ERα and ERβ) and arginine vasopressin (AVP), delving into their significant roles in modulating empathy, a critical psychological component in human social dynamics. Empathy, integrating affective and cognitive elements, is anchored in neural regions like the amygdala and prefrontal cortex. ERα and ERβ, pivotal in estrogen regulation, influence neurotransmitter dynamics and neural network activities, crucial for empathic development. AVP, key in regulating water balance, blood pressure, and social behaviors, interplays with these receptors, profoundly impacting empathic responses. The study highlights that ERα predominantly enhances empathy, especially affective empathy, by stimulating AVP synthesis and release. In contrast, ERβ may diminish empathy in certain contexts by suppressing AVP expression and activity. The intricate interplay, homeostatic balance, and mutual conversion between ERα and ERβ in AVP regulation are identified as challenging yet crucial areas for future research. These findings provide essential insights into the neurobiological underpinnings of empathy, offering new avenues for therapeutic interventions in social cognitive disorders and emotional dysregulation.
Collapse
Affiliation(s)
- Rui Du
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Ting Liang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Guofang Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
3
|
Alewel DI, Kodavanti UP. Neuroendocrine contribution to sex-related variations in adverse air pollution health effects. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2024; 27:287-314. [PMID: 39075643 DOI: 10.1080/10937404.2024.2383637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
Air pollution exposure is ranked as a leading environmental risk factor for not only cardiopulmonary diseases but also for systemic health ailments including diabetes, reproductive abnormalities, and neuropsychiatric disorders, likely mediated by central neural stress mechanisms. Current experimental evidence links many air pollution health outcomes with activation of neuroendocrine sympathetic-adrenal-medullary and hypothalamic-pituitary-adrenal (HPA) stress axes associated with resultant increases in adrenal-derived hormone levels acting as circulating mediators of multi-organ stress reactions. Epidemiological and experimental investigations also demonstrated sex-specific responses to air pollutant inhalation, which may be attributed to hormonal interactions within the stress and reproductive axes. Sex hormones (androgens and estrogens) interact with neuroendocrine functions to influence hypothalamic responses, subsequently augmenting stress-mediated metabolic and immune changes. These neurohormonal interactions may contribute to innate sex-specific responses to inhaled irritants, inducing differing individual susceptibility. The aim of this review was to: (1) examine neuroendocrine co-regulation of the HPA axis by gonadal hormones, (2) provide experimental evidence demonstrating sex-specific respiratory and systemic effects attributed to air pollutant inhalation exposure, and (3) postulate proposed mechanisms of stress and sex hormone interactions during air pollution-related stress.
Collapse
Affiliation(s)
- Devin I Alewel
- Oak Ridge Institute for Science and Education Research Participation Program, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Urmila P Kodavanti
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| |
Collapse
|
4
|
Nishimura K, Ueta Y, Yoshino K. Estrogen-dependent oxytocin expression in the hypothalamus and estrogen-dependent vasopressin in the median eminence. J Obstet Gynaecol Res 2024; 50:2009-2018. [PMID: 39340151 DOI: 10.1111/jog.16100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024]
Abstract
The posterior pituitary (PP) hormones oxytocin (OXT) and arginine vasopressin (AVP) are synthesized within the hypothalamic nucleus and released from the PP into systemic circulation. Hypothalamic AVP projects its axons into the external layer of median eminence (eME) and regulates anterior pituitary hormone secretion during stress responses. Although similar as PP hormones, we demonstrate distinct regulatory roles of estrogen in hypothalamic OXT and AVP dynamics. OXT dynamics in the hypothalamus exhibit sex-dependent variations and that estrogen may influence dynamic OXT level changes, as observed in OXT-mRFP1 transgenic rats. Estrogen was also observed to modulate dynamic changes in AVP levels in the axon terminals of eME in female AVP-eGFP transgenic rats. Although OXT and AVP are produced within the similar hypothalamic region, both exhibit distinct dynamics within the hypothalamus. Estrogen acts on the hypothalamus, and further effects of estrogen replacement therapy can be expected.
Collapse
Affiliation(s)
- Kazuaki Nishimura
- Department of Obstetrics and Gynecology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kiyoshi Yoshino
- Department of Obstetrics and Gynecology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
5
|
Zuloaga DG, Lafrican JJ, Zuloaga KL. Androgen regulation of behavioral stress responses and the hypothalamic-pituitary-adrenal axis. Horm Behav 2024; 162:105528. [PMID: 38503191 PMCID: PMC11144109 DOI: 10.1016/j.yhbeh.2024.105528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/02/2024] [Accepted: 03/06/2024] [Indexed: 03/21/2024]
Abstract
Testosterone is a powerful steroid hormone that can impact the brain and behavior in various ways, including regulating behavioral and neuroendocrine (hypothalamic-pituitary-adrenal (HPA) axis) stress responses. Early in life androgens can act to alter development of brain regions associated with stress regulation, which ultimately impacts the display of stress responses later in life. Adult circulating androgens can also influence the expression of distinct genes and proteins that regulate stress responses. These changes in the brain are hypothesized to underlie the potent effects of androgens in regulating behaviors related to stress and stress-induced activation of the HPA axis. Androgens can induce alterations in these functions through direct binding to the androgen receptor (AR) or following conversion to estrogens and subsequent binding to estrogen receptors including estrogen receptor alpha (ERα), beta (ERβ), and G protein-coupled estrogen receptor 1 (GPER1). In this review, we focus on the role of androgens in regulating behavioral and neuroendocrine stress responses at different stages of the lifespan and the sex hormone receptors involved in regulating these effects. We also review the specific brain regions and cell phenotypes upon which androgens are proposed to act to regulate stress responses with an emphasis on hypothalamic and extended amygdala subregions. This knowledge of androgen effects on these neural systems is critical for understanding how sex hormones regulate stress responses.
Collapse
Affiliation(s)
- Damian G Zuloaga
- Department of Psychology, University at Albany, Albany, NY, USA.
| | | | - Kristen L Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| |
Collapse
|
6
|
Gharibi S, Vaillend C, Lindsay A. The unconditioned fear response in vertebrates deficient in dystrophin. Prog Neurobiol 2024; 235:102590. [PMID: 38484964 DOI: 10.1016/j.pneurobio.2024.102590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/31/2024] [Accepted: 03/05/2024] [Indexed: 03/19/2024]
Abstract
Dystrophin loss due to mutations in the Duchenne muscular dystrophy (DMD) gene is associated with a wide spectrum of neurocognitive comorbidities, including an aberrant unconditioned fear response to stressful/threat stimuli. Dystrophin-deficient animal models of DMD demonstrate enhanced stress reactivity that manifests as sustained periods of immobility. When the threat is repetitive or severe in nature, dystrophinopathy phenotypes can be exacerbated and even cause sudden death. Thus, it is apparent that enhanced sensitivity to stressful/threat stimuli in dystrophin-deficient vertebrates is a legitimate cause of concern for patients with DMD that could impact neurocognition and pathophysiology. This review discusses our current understanding of the mechanisms and consequences of the hypersensitive fear response in preclinical models of DMD and the potential challenges facing clinical translatability.
Collapse
Affiliation(s)
- Saba Gharibi
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Cyrille Vaillend
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, Saclay 91400, France.
| | - Angus Lindsay
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia; School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand; Department of Medicine, University of Otago, Christchurch 8014, New Zealand.
| |
Collapse
|
7
|
Booth AM, Viernes R, Farrar VS, Flores L, Austin SH, Calisi RM. Sex-specific behavioral and physiological changes during single parenting in a biparental species, Columba livia. Horm Behav 2023; 156:105428. [PMID: 37748275 DOI: 10.1016/j.yhbeh.2023.105428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/27/2023]
Abstract
Many species exhibit biparental care to maximize fitness. When a partner is lost, the surviving partner may alter their behavior to compensate offspring. Whether both sexes use the same physiological mechanisms to manifest their change in behavior remains elusive. We investigated behaviors and mechanisms associated with the alteration of parental care post-partner removal in a biparental avian species, the rock dove (Columba livia). We hypothesized that rock dove single parents experience sex-biased changes in neural genomic transcription and reproductive behaviors, and these changes are related to chick development. We manipulated parental partner presence and measured parental attendance, offspring growth, gene expression of glucocorticoid receptors (GR) and mineralocorticoid receptors (MR) in the pituitary, and GR, MR, and estrogen receptor beta (ER-β) in the hypothalamus. We also measured circulating plasma concentrations of the stress-associated hormone corticosterone and the parental care-associated hormone prolactin. We also quantified prolactin gene (PRL) expression changes in the pituitary, as well as prolactin receptor (PRLR) expression in the hypothalamus and pituitary. We found that single mothers and fathers maintained similar provisioning levels as paired parents, but spent less cumulative time brooding chicks. Chicks of single parents were smaller than paired-parented chicks after three days post-hatch. Mothers in both treatment groups experienced higher expression of hypothalamic GR as compared to fathers. Single parents experienced lower PRL gene expression in the pituitary as compared to paired parents. No significant differences were found for the circulating hormones or other genes listed.
Collapse
Affiliation(s)
- April M Booth
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States of America.
| | - Rechelle Viernes
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States of America
| | - Victoria S Farrar
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States of America
| | - Laura Flores
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States of America
| | - Suzanne H Austin
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States of America
| | - Rebecca M Calisi
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States of America
| |
Collapse
|
8
|
Rybka KA, Lafrican JJ, Rosinger ZJ, Ariyibi DO, Brooks MR, Jacobskind JS, Zuloaga DG. Sex differences in androgen receptor, estrogen receptor alpha, and c-Fos co-expression with corticotropin releasing factor expressing neurons in restrained adult mice. Horm Behav 2023; 156:105448. [PMID: 38344954 PMCID: PMC10861933 DOI: 10.1016/j.yhbeh.2023.105448] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/03/2023] [Accepted: 10/23/2023] [Indexed: 02/15/2024]
Abstract
Gonadal hormone actions through androgen receptor (AR) and estrogen receptor alpha (ERα) regulate sex differences in hypothalamic-pituitary-adrenal (HPA) axis responsivity and stress-related behaviors. Here we tested whether corticotropin releasing factor (CRF) expressing neurons, which are widely known to regulate neuroendocrine and behavioral stress responses, co-express AR and ERα as a potential mechanism for gonadal hormone regulation of these responses. Using Crh-IRES-Cre::Ai9 reporter mice we report high co-localization of AR in CRF neurons within the medial preoptic area (MPOA), bed nucleus of the stria terminalis (BST), medial amygdala (MeA), and ventromedial hypothalamus (VMH), moderate levels within the central amygdala (CeA) and low levels in the paraventricular hypothalamus (PVN). Sex differences in CRF/AR co-expression were found in the principal nucleus of the BST (BSTmpl), CeA, MeA, and VMH (males>females). CRF co-localization with ERα was generally lower relative to AR co-localization. However, high co-expression was found within the MPOA, AVPV, and VMH, with moderate co-expression in the arcuate nucleus (ARC), BST, and MeA and low levels in the PVN and CeA. Sex differences in CRF/ERα co-localization were found in the BSTmpl and PVN (males>females). Finally, we assessed neural activation of CRF neurons in restraint-stressed mice and found greater CRF/c-Fos co-expression in females in the BSTmpl and periaqueductal gray, while co-expression was higher in males within the ARC and dorsal CA1. Given the known role of CRF in regulating behavioral stress responses and the HPA axis, AR/ERα co-expression and sex-specific activation of CRF cell groups indicate potential mechanisms for modulating sex differences in these functions.
Collapse
Affiliation(s)
- Krystyna A Rybka
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States of America
| | - Jennifer J Lafrican
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States of America
| | - Zachary J Rosinger
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States of America
| | - Deborah O Ariyibi
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States of America
| | - Mecca R Brooks
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States of America
| | - Jason S Jacobskind
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States of America
| | - Damian G Zuloaga
- Department of Psychology, University at Albany, State University New York, 1400 Washington Avenue, Albany, NY 12222, United States of America.
| |
Collapse
|
9
|
Raise-Abdullahi P, Meamar M, Vafaei AA, Alizadeh M, Dadkhah M, Shafia S, Ghalandari-Shamami M, Naderian R, Afshin Samaei S, Rashidy-Pour A. Hypothalamus and Post-Traumatic Stress Disorder: A Review. Brain Sci 2023; 13:1010. [PMID: 37508942 PMCID: PMC10377115 DOI: 10.3390/brainsci13071010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
Humans have lived in a dynamic environment fraught with potential dangers for thousands of years. While fear and stress were crucial for the survival of our ancestors, today, they are mostly considered harmful factors, threatening both our physical and mental health. Trauma is a highly stressful, often life-threatening event or a series of events, such as sexual assault, war, natural disasters, burns, and car accidents. Trauma can cause pathological metaplasticity, leading to long-lasting behavioral changes and impairing an individual's ability to cope with future challenges. If an individual is vulnerable, a tremendously traumatic event may result in post-traumatic stress disorder (PTSD). The hypothalamus is critical in initiating hormonal responses to stressful stimuli via the hypothalamic-pituitary-adrenal (HPA) axis. Linked to the prefrontal cortex and limbic structures, especially the amygdala and hippocampus, the hypothalamus acts as a central hub, integrating physiological aspects of the stress response. Consequently, the hypothalamic functions have been attributed to the pathophysiology of PTSD. However, apart from the well-known role of the HPA axis, the hypothalamus may also play different roles in the development of PTSD through other pathways, including the hypothalamic-pituitary-thyroid (HPT) and hypothalamic-pituitary-gonadal (HPG) axes, as well as by secreting growth hormone, prolactin, dopamine, and oxytocin. This review aims to summarize the current evidence regarding the neuroendocrine functions of the hypothalamus, which are correlated with the development of PTSD. A better understanding of the role of the hypothalamus in PTSD could help develop better treatments for this debilitating condition.
Collapse
Affiliation(s)
| | - Morvarid Meamar
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Abbas Ali Vafaei
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
- Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maryam Alizadeh
- Department of Basic Medical Sciences, Faculty of Medicine, Qom Medical Sciences, Islamic Azad University, Qom, Iran
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Sakineh Shafia
- Immunogenetics Research Center, Department of Physiology, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Ramtin Naderian
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Seyed Afshin Samaei
- Department of Neurology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
- Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
10
|
Acharya KD, Graham M, Raman H, Parakoyi AER, Corcoran A, Belete M, Ramaswamy B, Koul S, Sachar I, Derendorf K, Wilmer JB, Gottipati S, Tetel MJ. Estradiol-mediated protection against high-fat diet induced anxiety and obesity is associated with changes in the gut microbiota in female mice. Sci Rep 2023; 13:4776. [PMID: 36959275 PMCID: PMC10036463 DOI: 10.1038/s41598-023-31783-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/17/2023] [Indexed: 03/25/2023] Open
Abstract
Decreased estrogens during menopause are associated with increased risk of anxiety, depression, type 2 diabetes and obesity. Similarly, depleting estrogens in rodents by ovariectomy, combined with a high-fat diet (HFD), increases anxiety and adiposity. How estrogens and diet interact to affect anxiety and metabolism is poorly understood. Mounting evidence indicates that gut microbiota influence anxiety and metabolism. Here, we investigated the effects of estradiol (E) and HFD on anxiety, metabolism, and their correlation with changes in gut microbiota in female mice. Adult C57BL/6J mice were ovariectomized, implanted with E or vehicle-containing capsules and fed a standard diet or HFD. Anxiety-like behavior was assessed and neuronal activation was measured by c-fos immunoreactivity throughout the brain using iDISCO. HFD increased anxiety-like behavior, while E reduced this HFD-dependent anxiogenic effect. Interestingly, E decreased neuronal activation in brain regions involved in anxiety and metabolism. E treatment also altered gut microbes, a subset of which were associated with anxiety-like behavior. These findings provide insight into gut microbiota-based therapies for anxiety and metabolic disorders associated with declining estrogens in menopausal women.
Collapse
Affiliation(s)
- Kalpana D Acharya
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | - Madeline Graham
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | - Harshini Raman
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | | | - Alexis Corcoran
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | - Merzu Belete
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | - Bharath Ramaswamy
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | - Shashikant Koul
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | | | - Kevin Derendorf
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | - Jeremy B Wilmer
- Department of Psychology, Wellesley College, Wellesley, MA, 02481, USA
| | - Srikanth Gottipati
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, 08540, USA
| | - Marc J Tetel
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA.
| |
Collapse
|
11
|
Power EM, Ganeshan D, Iremonger KJ. Estradiol regulates voltage-gated potassium currents in corticotropin-releasing hormone neurons. J Exp Biol 2023; 226:287072. [PMID: 36805713 PMCID: PMC10038157 DOI: 10.1242/jeb.245222] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/08/2023] [Indexed: 02/22/2023]
Abstract
Corticotropin-releasing hormone (CRH) neurons are the primary neural population controlling the hypothalamic-pituitary-adrenal (HPA) axis and the secretion of adrenal stress hormones. Previous work has demonstrated that stress hormone secretion can be regulated by circulating levels of estradiol. However, the effect of estradiol on CRH neuron excitability is less clear. Here, we show that chronic estradiol replacement following ovariectomy increases two types of potassium channel currents in CRH neurons: fast inactivating voltage-gated A-type K+ channel currents (IA) and non-inactivating M-type K+ channel currents (IM). Despite the increase in K+ currents following estradiol replacement, there was no overall change in CRH neuron spiking excitability assessed with either frequency-current curves or current ramps. Together, these data reveal a complex picture whereby ovariectomy and estradiol replacement differentially modulate distinct aspects of CRH neuron and HPA axis function.
Collapse
Affiliation(s)
- Emmet M Power
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin9016, New Zealand
| | - Dharshini Ganeshan
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin9016, New Zealand
| | - Karl J Iremonger
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin9016, New Zealand
| |
Collapse
|
12
|
Smiley CE, Pate BS, Bouknight SJ, Francis MJ, Nowicki AV, Harrington EN, Wood SK. Estrogen receptor beta in the central amygdala regulates the deleterious behavioral and neuronal consequences of repeated social stress in female rats. Neurobiol Stress 2023; 23:100531. [PMID: 36879670 PMCID: PMC9984877 DOI: 10.1016/j.ynstr.2023.100531] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/02/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
While over 95% of the population has reported experiencing extreme stress or trauma, females of reproductive age develop stress-induced neuropsychiatric disorders at twice the rate of males. This suggests that ovarian hormones may facilitate neural processes that increase stress susceptibility and underlie the heightened rates of these disorders, like depression and anxiety, that result from stress exposure in females. However, there is contradicting evidence in the literature regarding estrogen's role in stress-related behavioral outcomes. Estrogen signaling through estrogen receptor beta (ERβ) has been traditionally thought of as anxiolytic, but recent studies suggest estrogen exhibits distinct effects in the context of stress. Furthermore, ERβ is found abundantly in many stress-sensitive brain loci, including the central amygdala (CeA), in which transcription of the vital stress hormone, corticotropin releasing factor (CRF), can be regulated by an estrogen response element. Therefore, these experiments sought to identify the role of CeA ERβ activity during stress on behavioral outcomes in naturally cycling, adult, female Sprague-Dawley rats. Rats were exposed to an ethological model of vicarious social stress, witness stress (WS), in which they experienced the sensory and psychological aspects of an aggressive social defeat encounter between two males. Following WS, rats exhibited stress-induced anxiety-like behaviors in the marble burying taskand brain analysis revealed increased ERβ and CRF specifically within the CeA following exposure to stress cues. Subsequent experiments were designed to target this receptor in the CeA using microinjections of the ERβ antagonist, PHTPP, prior to each stress session. During WS, estrogen signaling through ERβ was responsible for the behavioral sensitization to repeated social stress. Sucrose preference, acoustic startle, and marble burying tasks determined that blocking ERβ in the CeA during WS prevented the development of depressive-, anxiety-like, and hypervigilant behaviors. Additionally, brain analysis revealed a long-term decrease of intra-CeA CRF expression in PHTPP-treated rats. These experiments indicate that ERβ signaling in the CeA, likely through its effects on CRF, contributes to the development of negative valence behaviors that result from exposure to repeated social stress in female rats.
Collapse
Key Words
- ACTH, adrenocorticotropic hormone
- ASR, acoustic startle responding
- Anxiety
- BCA, bicinchoninic acid
- CON, control handing
- CORT, corticosterone
- CRF, corticotropin releasing factor
- CeA, central amygdala
- Central amygdala
- Corticotropin releasing factor
- EPM, elevated plus maze
- ERβ, estrogen receptor beta
- Estrogen receptor beta
- HPA, hypothalamic pituitary adrenal axis
- LC, locus coeruleus
- MB, marble burying
- PHTPP, 4-[2-Phenyl-5: 7-bis (trifluoromethyl) pyrazolo [1,5-a] pyrimidine-3- yl] phenol
- SPT, sucrose preference testing
- Social stress
- WS, witness stress
- dB, decibels
Collapse
Affiliation(s)
- Cora E. Smiley
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
- Dorn VA Medical Center, Columbia, SC, USA
| | - Brittany S. Pate
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
- University of South Carolina, Arnold School of Public Health, Department of Exercise Science, Columbia, SC, USA
| | - Samantha J. Bouknight
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
| | - Megan J. Francis
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
| | - Alexandria V. Nowicki
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
| | - Evelynn N. Harrington
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
| | - Susan K. Wood
- University of South Carolina, School of Medicine, Department of Pharmacology Physiology and Neuroscience, Columbia, SC, USA
- Dorn VA Medical Center, Columbia, SC, USA
| |
Collapse
|
13
|
He J, Yan JJ, Zha X, Ding XJ, Zhang YL, Lu Z, Xu XH. Sexually dimorphic effects of estrogen receptor 2 deletion in the dorsal raphe nucleus on emotional behaviors. J Neuroendocrinol 2023; 35:e13195. [PMID: 36072992 DOI: 10.1111/jne.13195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 08/03/2022] [Accepted: 08/16/2022] [Indexed: 11/27/2022]
Abstract
Sex differences in emotional behaviors and affective disorders have been widely noted, of which sexually dimorphic secretion of gonadal steroid hormones such as estrogen is suspected to play a role. However, the underlying neural mechanisms remain poorly understood. We noted that the expression of estrogen receptor 2 (Esr2, or ERβ), a key mediator of estrogen signaling in the brain, was enriched in the dorsal raphe nucleus (DRN), a region involved in emotion regulation. To investigate whether DRN Esr2 expression confers sex-specific susceptibility or vulnerability in emotional behaviors, we generated a conditional allele of Esr2 that allowed for site-specific deletion of Esr2 in the DRN via local injection of Cre-expressing viruses. DRN-specific Esr2 deletion mildly increased anxiety behaviors in females, as shown by decreased time spent in the center zone of an open field in knockout females. By contrast, DRN Esr2 deletion had no effects on anxiety levels in males, as demonstrated by knockout males spending comparable time in the center zone of an open field and open arms of an elevated-plus maze. Furthermore, in the tail suspension test, DRN Esr2 deletion reduced immobility, a depression-like behavior, in a male-biased manner. Together, these results reveal sex-specific functions of DRN Esr2 in regulating emotional behaviors and suggest targeted manipulation of DRN Esr2 signaling as a potential therapeutic strategy to treat sex-biased affective disorders.
Collapse
Affiliation(s)
- Jing He
- Department of Psychiatry, Tongji Hospital of Tongji University, Tongji University School of Medicine, Shanghai, China
| | - Jing-Jing Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Xi Zha
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Xiao-Jing Ding
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Yan-Li Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Zheng Lu
- Department of Psychiatry, Tongji Hospital of Tongji University, Tongji University School of Medicine, Shanghai, China
| | - Xiao-Hong Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| |
Collapse
|
14
|
Helman TJ, Headrick JP, Stapelberg NJC, Braidy N. The sex-dependent response to psychosocial stress and ischaemic heart disease. Front Cardiovasc Med 2023; 10:1072042. [PMID: 37153459 PMCID: PMC10160413 DOI: 10.3389/fcvm.2023.1072042] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Stress is an important risk factor for modern chronic diseases, with distinct influences in males and females. The sex specificity of the mammalian stress response contributes to the sex-dependent development and impacts of coronary artery disease (CAD). Compared to men, women appear to have greater susceptibility to chronic forms of psychosocial stress, extending beyond an increased incidence of mood disorders to include a 2- to 4-fold higher risk of stress-dependent myocardial infarction in women, and up to 10-fold higher risk of Takotsubo syndrome-a stress-dependent coronary-myocardial disorder most prevalent in post-menopausal women. Sex differences arise at all levels of the stress response: from initial perception of stress to behavioural, cognitive, and affective responses and longer-term disease outcomes. These fundamental differences involve interactions between chromosomal and gonadal determinants, (mal)adaptive epigenetic modulation across the lifespan (particularly in early life), and the extrinsic influences of socio-cultural, economic, and environmental factors. Pre-clinical investigations of biological mechanisms support distinct early life programming and a heightened corticolimbic-noradrenaline-neuroinflammatory reactivity in females vs. males, among implicated determinants of the chronic stress response. Unravelling the intrinsic molecular, cellular and systems biological basis of these differences, and their interactions with external lifestyle/socio-cultural determinants, can guide preventative and therapeutic strategies to better target coronary heart disease in a tailored sex-specific manner.
Collapse
Affiliation(s)
- Tessa J. Helman
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, NSW, Sydney, Australia
- Correspondence: Tessa J. Helman
| | - John P. Headrick
- Schoolof Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
| | | | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, NSW, Sydney, Australia
| |
Collapse
|
15
|
Hokenson RE, Alam YH, Short AK, Jung S, Jang C, Baram TZ. Sex-dependent effects of multiple acute concurrent stresses on memory: a role for hippocampal estrogens. Front Behav Neurosci 2022; 16:984494. [PMID: 36160685 PMCID: PMC9492881 DOI: 10.3389/fnbeh.2022.984494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/16/2022] [Indexed: 11/26/2022] Open
Abstract
Memory disruption commonly follows chronic stress, whereas acute stressors are generally benign. However, acute traumas such as mass shootings or natural disasters—lasting minutes to hours and consisting of simultaneous physical, social, and emotional stresses—are increasingly recognized as significant risk factors for memory problems and PTSD. Our prior work has revealed that these complex stresses (concurrent multiple acute stresses: MAS) disrupt hippocampus-dependent memory in male rodents. In females, the impacts of MAS are estrous cycle-dependent: MAS impairs memory during early proestrus (high estrogens phase), whereas the memory of female mice stressed during estrus (low estrogens phase) is protected. Female memory impairments limited to high estrogens phases suggest that higher levels of estrogens are necessary for MAS to disrupt memory, supported by evidence that males have higher hippocampal estradiol than estrous females. To test the role of estrogens in stress-induced memory deficits, we blocked estrogen production using aromatase inhibitors. A week of blockade protected male and female mice from MAS-induced memory disturbances, suggesting that high levels of estrogens are required for stress-provoked memory impairments in both males and females. To directly quantify 17β-estradiol in murine hippocampus we employed both ELISA and mass spectrometry and identified significant confounders in both procedures. Taken together, the cross-cycle and aromatase studies in males and females support the role for high hippocampal estrogens in mediating the effect of complex acute stress on memory. Future studies focus on the receptors involved, the longevity of these effects, and their relation to PTSD-like behaviors in experimental models.
Collapse
Affiliation(s)
- Rachael E. Hokenson
- Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, United States
- *Correspondence: Rachael E. Hokenson
| | - Yasmine H. Alam
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States
| | - Annabel K. Short
- Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, United States
- Department of Pediatrics, University of California, Irvine, Irvine, CA =, United States
| | - Sunhee Jung
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States
| | - Tallie Z. Baram
- Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, United States
- Department of Pediatrics, University of California, Irvine, Irvine, CA =, United States
- Department of Neurology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
16
|
Handa RJ, Sheng JA, Castellanos EA, Templeton HN, McGivern RF. Sex Differences in Acute Neuroendocrine Responses to Stressors in Rodents and Humans. Cold Spring Harb Perspect Biol 2022; 14:a039081. [PMID: 35667789 PMCID: PMC9438783 DOI: 10.1101/cshperspect.a039081] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Sex differences in the neuroendocrine response to acute stress occur in both animals and humans. In rodents, stressors such as restraint and novelty induce a greater activation of the hypothalamic-pituitary-adrenal axis (HPA) in females compared to males. The nature of this difference arises from steroid actions during development (organizational effects) and adulthood (activational effects). Androgens decrease HPA stress responsivity to acute stress, while estradiol increases it. Androgenic down-regulation of HPA responsiveness is mediated by the binding of testosterone (T) and dihydrotestosterone (DHT) to the androgen receptor, as well as the binding of the DHT metabolite, 3β-diol, to the β form of the estrogen receptor (ERβ). Estradiol binding to the α form of the estrogen receptor (ERα) increases HPA responsivity. Studies of human sex differences are relatively few and generally employ a psychosocial paradigm to measure stress-related HPA activation. Men consistently show greater HPA reactivity than women when being evaluated for achievement. Some studies have found greater reactivity in women when being evaluated for social performance. The pattern is inconsistent with rodent studies but may involve the differential nature of the stressors employed. Psychosocial stress is nonphysical and invokes a significant degree of top-down processing that is not easily comparable to the types of stressors employed in rodents. Gender identity may also be a factor based on recent work showing that it influences the neural processing of positive and negative emotional stimuli independent of genetic sex. Comparing different types of stressors and how they interact with gender identity and genetic sex will provide a better understanding of sex steroid influences on stress-related HPA reactivity.
Collapse
Affiliation(s)
- Robert J Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Julietta A Sheng
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Emily A Castellanos
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Hayley N Templeton
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Robert F McGivern
- Department of Psychology, San Diego State University, San Diego, California 92120, USA
| |
Collapse
|
17
|
An X, Guo W, Wu H, Fu X, Li M, Zhang Y, Li Y, Cui R, Yang W, Zhang Z, Zhao G. Sex Differences in Depression Caused by Early Life Stress and Related Mechanisms. Front Neurosci 2022; 16:797755. [PMID: 35663561 PMCID: PMC9157793 DOI: 10.3389/fnins.2022.797755] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/02/2022] [Indexed: 11/26/2022] Open
Abstract
Depression is a common psychiatric disease caused by various factors, manifesting with continuous low spirits, with its precise mechanism being unclear. Early life stress (ELS) is receiving more attention as a possible cause of depression. Many studies focused on the mechanisms underlying how ELS leads to changes in sex hormones, neurotransmitters, hypothalamic pituitary adrenocortical (HPA) axis function, and epigenetics. The adverse effects of ELS on adulthood are mainly dependent on the time window when stress occurs, sex and the developmental stage when evaluating the impacts. Therefore, with regard to the exact sex differences of adult depression, we found that ELS could lead to sex-differentiated depression through multiple mechanisms, including 5-HT, sex hormone, HPA axis, and epigenetics.
Collapse
Affiliation(s)
- Xianquan An
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Department of Anesthesiology, Second Hospital of Jilin University, Changchun, China
| | - Wanxu Guo
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Huiying Wu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Xiying Fu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Ming Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Yizhi Zhang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Yanlin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Zhuo Zhang
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
- *Correspondence: Zhuo Zhang,
| | - Guoqing Zhao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Guoqing Zhao,
| |
Collapse
|
18
|
Rybka KA, Sturm KL, De Guzman RM, Bah S, Jacobskind JS, Rosinger ZJ, Taroc EZM, Forni PE, Zuloaga DG. Androgen regulation of corticotropin releasing factor receptor 1 in the mouse brain. Neuroscience 2022; 491:185-199. [DOI: 10.1016/j.neuroscience.2022.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 12/19/2022]
|
19
|
Grigoryan GA. Ovariectomy as a Model of Anxiety-Depressive Disorders. NEUROCHEM J+ 2022. [DOI: 10.1134/s1819712422010068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Moran CA, Collins LF, Beydoun N, Mehta PK, Fatade Y, Isiadinso I, Lewis TT, Weber B, Goldstein J, Ofotokun I, Quyyumi A, Choi MY, Titanji K, Lahiri CD. Cardiovascular Implications of Immune Disorders in Women. Circ Res 2022; 130:593-610. [PMID: 35175848 PMCID: PMC8869407 DOI: 10.1161/circresaha.121.319877] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immune responses differ between men and women, with women at higher risk of developing chronic autoimmune diseases and having more robust immune responses to many viruses, including HIV and hepatitis C virus. Although immune dysregulation plays a prominent role in chronic systemic inflammation, a key driver in the development of atherosclerotic cardiovascular disease (ASCVD), standard ASCVD risk prediction scores underestimate risk in populations with immune disorders, particularly women. This review focuses on the ASCVD implications of immune dysregulation due to disorders with varying global prevalence by sex: autoimmune disorders (female predominant), HIV (male-female equivalent), and hepatitis C virus (male predominant). Factors contributing to ASCVD in women with immune disorders, including traditional risk factors, dysregulated innate and adaptive immunity, sex hormones, and treatment modalities, are discussed. Finally, the need to develop new ASCVD risk stratification tools that incorporate variables specific to populations with chronic immune disorders, particularly in women, is emphasized.
Collapse
Affiliation(s)
- Caitlin A. Moran
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, GA, USA
| | - Lauren F. Collins
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, GA, USA
| | - Nour Beydoun
- Emory University School of Medicine, Department of Medicine, Center for Heart Disease Prevention, Division of Cardiology and Emory Women’s Heart Center, Atlanta, GA, USA
| | - Puja K. Mehta
- Emory University School of Medicine, Department of Medicine, Center for Heart Disease Prevention, Division of Cardiology and Emory Women’s Heart Center, Atlanta, GA, USA
| | - Yetunde Fatade
- Emory University School of Medicine, Department of Medicine, Atlanta, GA, USA
| | - Ijeoma Isiadinso
- Emory University School of Medicine, Department of Medicine, Center for Heart Disease Prevention, Division of Cardiology and Emory Women’s Heart Center, Atlanta, GA, USA
| | - Tené T Lewis
- Emory University, Rollins School of Public Health, Department of Epidemiology, Atlanta, GA, USA
| | - Brittany Weber
- Harvard Medical School, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Jill Goldstein
- Massachusetts General Hospital, Department of Psychiatry, and Harvard Medical School, Departments of Psychiatry and Medicine, Boston, MA, USA
| | - Igho Ofotokun
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, GA, USA
| | - Arshed Quyyumi
- Emory University School of Medicine, Department of Medicine, Center for Heart Disease Prevention, Division of Cardiology and Emory Women’s Heart Center, Atlanta, GA, USA
| | - May Y. Choi
- Cumming School of Medicine, University of Calgary, Calgary, AB Canada
| | - Kehmia Titanji
- Emory University, Department of Medicine, Division of Endocrinology, Metabolism, and Lipids, Atlanta, GA, USA
| | - Cecile D. Lahiri
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, GA, USA
| |
Collapse
|
21
|
Sheng JA, Tan SML, Hale TM, Handa RJ. Androgens and Their Role in Regulating Sex Differences in the Hypothalamic/Pituitary/Adrenal Axis Stress Response and Stress-Related Behaviors. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2022; 2:261-274. [PMID: 35024695 PMCID: PMC8744007 DOI: 10.1089/andro.2021.0021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/24/2021] [Indexed: 02/06/2023]
Abstract
Androgens play a pivotal role during development. These gonadal hormones and their receptors exert organizational actions that shape brain morphology in regions controlling the stress regulatory systems in a male-specific manner. Specifically, androgens drive sex differences in the hypothalamic/pituitary/adrenal (HPA) axis and corresponding hypothalamic neuropeptides. While studies have examined the role of estradiol and its receptors in sex differences in the HPA axis and associated behaviors, the role of androgens remains far less studied. Androgens are generally thought to modulate the HPA axis through the activation of androgen receptors (ARs). They can also impact the HPA axis through reduction to estrogenic metabolites that can bind estrogen receptors in the brain and periphery. Such regulation of the HPA axis stress response by androgens can often result in sex-biased risk factors for stress-related disorders, such as anxiety and depression. This review focuses on the biosynthesis pathways and molecular actions of androgens and their nuclear receptors. The impact of androgens on hypothalamic neuropeptide systems (corticotropin-releasing hormone, arginine vasopressin, oxytocin, dopamine, and serotonin) that control the stress response and stress-related disorders is discussed. Finally, this review discusses potential therapeutics involving androgens (androgen replacement therapies, selective AR modulator therapies) and ongoing clinical trials.
Collapse
Affiliation(s)
- Julietta A Sheng
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Sarah M L Tan
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Taben M Hale
- Department of Basic Medical Science, University of Arizona College of Medicine - Phoenix, Arizona, USA
| | - Robert J Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
22
|
Azcoitia I, Mendez P, Garcia-Segura LM. Aromatase in the Human Brain. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2021; 2:189-202. [PMID: 35024691 PMCID: PMC8744447 DOI: 10.1089/andro.2021.0007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 06/20/2021] [Indexed: 11/30/2022]
Abstract
The aromatase cytochrome P450 (P450arom) enzyme, or estrogen synthase, which is coded by the CYP19A1 gene, is widely expressed in a subpopulation of excitatory and inhibitory neurons, astrocytes, and other cell types in the human brain. Experimental studies in laboratory animals indicate a prominent role of brain aromatization of androgens to estrogens in regulating different brain functions. However, the consequences of aromatase expression in the human brain remain poorly understood. Here, we summarize the current knowledge about aromatase expression in the human brain, abundant in the thalamus, amygdala, hypothalamus, cortex, and hippocampus and discuss its role in the regulation of sensory integration, body homeostasis, social behavior, cognition, language, and integrative functions. Since brain aromatase is affected by neurodegenerative conditions and may participate in sex-specific manifestations of autism spectrum disorders, major depressive disorder, multiple sclerosis, stroke, and Alzheimer's disease, we discuss future avenues for research and potential clinical and therapeutic implications of the expression of aromatase in the human brain.
Collapse
Affiliation(s)
- Iñigo Azcoitia
- Department of Cell Biology, Faculty of Biology, Universidad Complutense de Madrid and Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Pablo Mendez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Luis M. Garcia-Segura
- Department of Cell Biology, Faculty of Biology, Universidad Complutense de Madrid and Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
23
|
Karamichos D, Escandon P, Vasini B, Nicholas SE, Van L, Dang DH, Cunningham RL, Riaz KM. Anterior pituitary, sex hormones, and keratoconus: Beyond traditional targets. Prog Retin Eye Res 2021; 88:101016. [PMID: 34740824 PMCID: PMC9058044 DOI: 10.1016/j.preteyeres.2021.101016] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/13/2022]
Abstract
"The Diseases of the Horny-coat of The Eye", known today as keratoconus, is a progressive, multifactorial, non-inflammatory ectatic corneal disorder that is characterized by steepening (bulging) and thinning of the cornea, irregular astigmatism, myopia, and scarring that can cause devastating vision loss. The significant socioeconomic impact of the disease is immeasurable, as patients with keratoconus can have difficulties securing certain jobs or even joining the military. Despite the introduction of corneal crosslinking and improvements in scleral contact lens designs, corneal transplants remain the main surgical intervention for treating keratoconus refractory to medical therapy and visual rehabilitation. To-date, the etiology and pathogenesis of keratoconus remains unclear. Research studies have increased exponentially over the years, highlighting the clinical significance and international interest in this disease. Hormonal imbalances have been linked to keratoconus, both clinically and experimentally, with both sexes affected. However, it is unclear how (molecular/cellular signaling) or when (age/disease stage(s)) those hormones affect the keratoconic cornea. Previous studies have categorized the human cornea as an extragonadal tissue, showing modulation of the gonadotropins, specifically luteinizing hormone (LH) and follicle-stimulating hormone (FSH). Studies herein provide new data (both in vitro and in vivo) to further delineate the role of hormones/gonadotropins in the keratoconus pathobiology, and propose the existence of a new axis named the Hypothalamic-Pituitary-Adrenal-Corneal (HPAC) axis.
Collapse
Affiliation(s)
- Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA.
| | - Paulina Escandon
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Brenda Vasini
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Sarah E Nicholas
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Lyly Van
- University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA; Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Deanna H Dang
- College of Medicine, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Kamran M Riaz
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
24
|
Involvement of Ghrelin Dynamics in Stress-Induced Eating Disorder: Effects of Sex and Aging. Int J Mol Sci 2021; 22:ijms222111695. [PMID: 34769125 PMCID: PMC8583769 DOI: 10.3390/ijms222111695] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Stress, a factor that affects appetite in our daily lives, enhances or suppresses appetite and changes palatability. However, so far, the mechanisms underlying the link between stress and eating have not been fully elucidated. Among the peripherally produced appetite-related peptides, ghrelin is the only orexigenic peptide, and abnormalities in the dynamics and reactivity of this peptide are involved in appetite abnormalities in various diseases and psychological states. This review presents an overview of the research results of studies evaluating the effects of various stresses on appetite. The first half of this review describes the relationship between appetite and stress, and the second half describes the relationship between the appetite-promoting peptide ghrelin and stress. The effects of sex differences and aging under stress on appetite are also described.
Collapse
|
25
|
Guerrieri GM, Ben Dor R, Li X, Wei SM, Martinez PE, Neiman LK, Rubinow DR, Schmidt PJ. The Cortisol and ACTH Response to Dex/CRH Testing in Women With and Without Perimenopausal Depression. J Clin Endocrinol Metab 2021; 106:3007-3018. [PMID: 34097071 PMCID: PMC8475231 DOI: 10.1210/clinem/dgab407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Indexed: 12/20/2022]
Abstract
CONTEXT Abnormalities in the hypothalamic-pituitary-adrenal (HPA) axis are frequent accompaniments of depression, and studies have documented the role of stress and stressful life events in the ontogeny of perimenopausal depressions (PMD). Because HPA axis function in women is further modulated both by aging and ovarian steroids, it is possible that a dysregulated HPA axis contributes to the increased risk of PMD. OBJECTIVE We examined HPA axis function in perimenopausal women with and without depression using the combined dexamethasone-corticotropin-releasing hormone (Dex/CRH) test. METHODS Dex/CRH tests were performed on 20 women with PMD and 20 women who were also perimenopausal but without current or past depression (control women). Main outcome measures were plasma levels of cortisol and adrenocorticotropin (ACTH) and 24-hour urinary free cortisol (UFC). Five women took chronic stable medications, otherwise all women were medically healthy, and both groups were comparable with respect to reproductive stage and age. Standardized symptom rating scales were administered to each woman prior to Dex/CRH testing. RESULTS No group differences were present in either baseline or stimulated ACTH and cortisol secretion. Baseline plasma measures of estradiol, progesterone, and 24-hour UFC levels similarly did not differ in PMD and control women. CONCLUSION Despite reports of increased stress responsiveness in PMD, we observed no abnormalities of HPA axis activity associated with PMD compared with women without depression. These findings suggest that PMD is not uniformly associated with HPA dysregulation and could reflect underlying pathophysiologic processes that are distinct from women with nonreproductive-related depressions.
Collapse
Affiliation(s)
- Gioia M Guerrieri
- Behavioral Endocrinology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Division of Anesthesiology, Addiction Medicine, and Pain Medicine, Office of Neuroscience, Food and Drug Administration, Silver Spring, MD, USA
| | - Rivka Ben Dor
- Behavioral Endocrinology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Xiaobai Li
- Biostatistics & Clinical Epidemiology Service, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Shau-Ming Wei
- Behavioral Endocrinology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Pedro E Martinez
- Behavioral Endocrinology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Lynnette K Neiman
- Program in Reproductive and Adult Endocrinology, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - David R Rubinow
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Peter J Schmidt
- Behavioral Endocrinology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Correspondence: Peter J. Schmidt, MD, National Institute of Mental Health, 10 CRC, Rm 25330, 10 Center Dr, MSC 1277, Bethesda, MD 20892-1277, USA.
| |
Collapse
|
26
|
Martynyuk AE, Ju LS, Morey TE. The potential role of stress and sex steroids in heritable effects of sevoflurane†. Biol Reprod 2021; 105:735-746. [PMID: 34192761 PMCID: PMC8444702 DOI: 10.1093/biolre/ioab129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/17/2021] [Accepted: 06/25/2021] [Indexed: 12/11/2022] Open
Abstract
Most surgical procedures require general anesthesia, which is a reversible deep sedation state lacking all perception. The induction of this state is possible because of complex molecular and neuronal network actions of general anesthetics (GAs) and other pharmacological agents. Laboratory and clinical studies indicate that the effects of GAs may not be completely reversible upon anesthesia withdrawal. The long-term neurocognitive effects of GAs, especially when administered at the extremes of ages, are an increasingly recognized health concern and the subject of extensive laboratory and clinical research. Initial studies in rodents suggest that the adverse effects of GAs, whose actions involve enhancement of GABA type A receptor activity (GABAergic GAs), can also extend to future unexposed offspring. Importantly, experimental findings show that GABAergic GAs may induce heritable effects when administered from the early postnatal period to at least young adulthood, covering nearly all age groups that may have children after exposure to anesthesia. More studies are needed to understand when and how the clinical use of GAs in a large and growing population of patients can result in lower resilience to diseases in the even larger population of their unexposed offspring. This minireview is focused on the authors' published results and data in the literature supporting the notion that GABAergic GAs, in particular sevoflurane, may upregulate systemic levels of stress and sex steroids and alter expressions of genes that are essential for the functioning of these steroid systems. The authors hypothesize that stress and sex steroids are involved in the mediation of sex-specific heritable effects of sevoflurane.
Collapse
Affiliation(s)
- Anatoly E Martynyuk
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL, USA
| | - Ling-Sha Ju
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Timothy E Morey
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
27
|
Martin EL, Doncheck EM, Reichel CM, McRae-Clark AL. Consideration of sex as a biological variable in the translation of pharmacotherapy for stress-associated drug seeking. Neurobiol Stress 2021; 15:100364. [PMID: 34345636 PMCID: PMC8319013 DOI: 10.1016/j.ynstr.2021.100364] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/23/2021] [Accepted: 07/08/2021] [Indexed: 12/01/2022] Open
Abstract
Stress is a frequent precipitant of relapse to drug use. Pharmacotherapies targeting a diverse array of neural systems have been assayed for efficacy in attenuating stress-induced drug-seeking in both rodents and in humans, but none have shown enough evidence of utility to warrant routine use in the clinic. We posit that a critical barrier in effective translation is inattention to sex as a biological variable at all phases of the research process. In this review, we detail the neurobiological systems implicated in stress-induced relapse to cocaine, opioids, methamphetamine, and cannabis, as well as the pharmacotherapies that have been used to target these systems in rodent models, the human laboratory, and in clinical trials. In each of these areas we additionally describe the potential influences of biological sex on outcomes, and how inattention to fundamental sex differences can lead to biases during drug development that contribute to the limited success of large clinical trials. Based on these observations, we determine that of the pharmacotherapies discussed only α2-adrenergic receptor agonists and oxytocin have a body of research with sufficient consideration of biological sex to warrant further clinical evaluation. Pharmacotherapies that target β-adrenergic receptors, other neuroactive peptides, the hypothalamic-pituitary-adrenal axis, neuroactive steroids, and the endogenous opioid and cannabinoid systems require further assessment in females at the preclinical and human laboratory levels before progression to clinical trials can be recommended.
Collapse
Affiliation(s)
- Erin L Martin
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Elizabeth M Doncheck
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Carmela M Reichel
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Aimee L McRae-Clark
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA.,Department of Psychiatry, Medical University of South Carolina, Charleston, SC, 29425, USA
| |
Collapse
|
28
|
Kawatake-Kuno A, Murai T, Uchida S. The Molecular Basis of Depression: Implications of Sex-Related Differences in Epigenetic Regulation. Front Mol Neurosci 2021; 14:708004. [PMID: 34276306 PMCID: PMC8282210 DOI: 10.3389/fnmol.2021.708004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/14/2021] [Indexed: 12/22/2022] Open
Abstract
Major depressive disorder (MDD) is a leading cause of disability worldwide. Although the etiology and pathophysiology of MDD remain poorly understood, aberrant neuroplasticity mediated by the epigenetic dysregulation of gene expression within the brain, which may occur due to genetic and environmental factors, may increase the risk of this disorder. Evidence has also been reported for sex-related differences in the pathophysiology of MDD, with female patients showing a greater severity of symptoms, higher degree of functional impairment, and more atypical depressive symptoms. Males and females also differ in their responsiveness to antidepressants. These clinical findings suggest that sex-dependent molecular and neural mechanisms may underlie the development of depression and the actions of antidepressant medications. This review discusses recent advances regarding the role of epigenetics in stress and depression. The first section presents a brief introduction of the basic mechanisms of epigenetic regulation, including histone modifications, DNA methylation, and non-coding RNAs. The second section reviews their contributions to neural plasticity, the risk of depression, and resilience against depression, with a particular focus on epigenetic modulators that have causal relationships with stress and depression in both clinical and animal studies. The third section highlights studies exploring sex-dependent epigenetic alterations associated with susceptibility to stress and depression. Finally, we discuss future directions to understand the etiology and pathophysiology of MDD, which would contribute to optimized and personalized therapy.
Collapse
Affiliation(s)
- Ayako Kawatake-Kuno
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshiya Murai
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shusaku Uchida
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
29
|
Moisan MP. Sexual Dimorphism in Glucocorticoid Stress Response. Int J Mol Sci 2021; 22:ijms22063139. [PMID: 33808655 PMCID: PMC8003420 DOI: 10.3390/ijms22063139] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/16/2021] [Indexed: 12/18/2022] Open
Abstract
Chronic stress is encountered in our everyday life and is thought to contribute to a number of diseases. Many of these stress-related disorders display a sex bias. Because glucocorticoid hormones are the main biological mediator of chronic stress, researchers have been interested in understanding the sexual dimorphism in glucocorticoid stress response to better explain the sex bias in stress-related diseases. Although not yet demonstrated for glucocorticoid regulation, sex chromosomes do influence sex-specific biology as soon as conception. Then a transient rise in testosterone start to shape the male brain during the prenatal period differently to the female brain. These organizational effects are completed just before puberty. The cerebral regions implicated in glucocorticoid regulation at rest and after stress are thereby impacted in a sex-specific manner. After puberty, the high levels of all gonadal hormones will interact with glucocorticoid hormones in specific crosstalk through their respective nuclear receptors. In addition, stress occurring early in life, in particular during the prenatal period and in adolescence will prime in the long-term glucocorticoid stress response through epigenetic mechanisms, again in a sex-specific manner. Altogether, various molecular mechanisms explain sex-specific glucocorticoid stress responses that do not exclude important gender effects in humans.
Collapse
|
30
|
Hokenson RE, Short AK, Chen Y, Pham AL, Adams ET, Bolton JL, Swarup V, Gall CM, Baram TZ. Unexpected Role of Physiological Estrogen in Acute Stress-Induced Memory Deficits. J Neurosci 2021; 41:648-662. [PMID: 33262247 PMCID: PMC7842761 DOI: 10.1523/jneurosci.2146-20.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 11/22/2022] Open
Abstract
Stress may promote emotional and cognitive disturbances, which differ by sex. Adverse outcomes, including memory disturbances, are typically observed following chronic stress, but are now being recognized also after short events, including mass shootings, assault, or natural disasters, events that consist of concurrent multiple acute stresses (MAS). Prior work has established profound and enduring effects of MAS on memory in males. Here we examined the effects of MAS on female mice and probed the role of hormonal fluctuations during the estrous cycle on MAS-induced memory problems and the underlying brain network and cellular mechanisms. Female mice were impacted by MAS in an estrous cycle-dependent manner: MAS impaired hippocampus-dependent spatial memory in early-proestrous mice, characterized by high levels of estradiol, whereas memory of mice stressed during estrus (low estradiol) was spared. As spatial memory requires an intact dorsal hippocampal CA1, we examined synaptic integrity in mice stressed at different cycle phases and found a congruence of dendritic spine density and spatial memory deficits, with reduced spine density only in mice stressed during high estradiol cycle phases. Assessing MAS-induced activation of brain networks interconnected with hippocampus, we identified differential estrous cycle-dependent activation of memory- and stress-related regions, including the amygdala. Network analyses of the cross-correlation of fos expression among these regions uncovered functional connectivity that differentiated impaired mice from those not impaired by MAS. In conclusion, the estrous cycle modulates the impact of MAS on spatial memory, and fluctuating physiological levels of sex hormones may contribute to this effect.SIGNIFICANCE STATEMENT: Effects of stress on brain functions, including memory, are profound and sex-dependent. Acute stressors occurring simultaneously result in spatial memory impairments in males, but effects on females are unknown. Here we identified estrous cycle-dependent effects of such stresses on memory in females. Surprisingly, females with higher physiological estradiol experienced stress-induced memory impairment and a loss of underlying synapses. Memory- and stress-responsive brain regions interconnected with hippocampus were differentially activated across high and low estradiol mice, and predicted memory impairment. Thus, at functional, network, and cellular levels, physiological estradiol influences the effects of stress on memory in females, providing insight into mechanisms of prominent sex differences in stress-related memory disorders, such as post-traumatic stress disorder.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Christine M Gall
- Departments of Anatomy and Neurobiology
- Neurobiology and Behavior
| | - Tallie Z Baram
- Departments of Anatomy and Neurobiology
- Pediatrics
- Neurology, University of California-Irvine, Irvine, California 92697
| |
Collapse
|
31
|
Sheng JA, Bales NJ, Myers SA, Bautista AI, Roueinfar M, Hale TM, Handa RJ. The Hypothalamic-Pituitary-Adrenal Axis: Development, Programming Actions of Hormones, and Maternal-Fetal Interactions. Front Behav Neurosci 2021; 14:601939. [PMID: 33519393 PMCID: PMC7838595 DOI: 10.3389/fnbeh.2020.601939] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
The hypothalamic-pituitary-adrenal axis is a complex system of neuroendocrine pathways and feedback loops that function to maintain physiological homeostasis. Abnormal development of the hypothalamic-pituitary-adrenal (HPA) axis can further result in long-term alterations in neuropeptide and neurotransmitter synthesis in the central nervous system, as well as glucocorticoid hormone synthesis in the periphery. Together, these changes can potentially lead to a disruption in neuroendocrine, behavioral, autonomic, and metabolic functions in adulthood. In this review, we will discuss the regulation of the HPA axis and its development. We will also examine the maternal-fetal hypothalamic-pituitary-adrenal axis and disruption of the normal fetal environment which becomes a major risk factor for many neurodevelopmental pathologies in adulthood, such as major depressive disorder, anxiety, schizophrenia, and others.
Collapse
Affiliation(s)
- Julietta A. Sheng
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Natalie J. Bales
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Sage A. Myers
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Anna I. Bautista
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Mina Roueinfar
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Taben M. Hale
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, United States
| | - Robert J. Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, United States
| |
Collapse
|
32
|
So SY, Savidge TC. Sex-Bias in Irritable Bowel Syndrome: Linking Steroids to the Gut-Brain Axis. Front Endocrinol (Lausanne) 2021; 12:684096. [PMID: 34093447 PMCID: PMC8170482 DOI: 10.3389/fendo.2021.684096] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder that is more common in females. Despite its high global incidence, the disease mechanism is still unclear and therapeutic options remain limited. The sexual dimorphism in IBS incidence suggests that sex steroids play a role in disease onset and symptoms severity. This review considers sex steroids and their involvement in IBS symptoms and the underlying disease mechanisms. Estrogens and androgens play important regulatory roles in IBS symptomology, including visceral sensitivity, gut motility and psychological conditions, possibly through modulating the gut-brain axis. Steroids are regulators of hypothalamic-pituitary-adrenal activity and autonomic nervous system function. They also modulate gut microbiota and enteric nervous systems, impacting serotonin and mast cell signaling. Sex steroids also facilitate bidirectional cross-talk between the microbiota and host following bacterial transformation and recycling of steroids by the intestine. The sex-specific interplay between sex steroids and the host provides neuroendocrinology insight into the pathophysiology, epigenetics and treatment of IBS patients.
Collapse
Affiliation(s)
- Sik Yu So
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Microbiome Center, Department of Pathology, Texas Children’s Hospital, Houston, TX, United States
| | - Tor C. Savidge
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Microbiome Center, Department of Pathology, Texas Children’s Hospital, Houston, TX, United States
- *Correspondence: Tor C. Savidge,
| |
Collapse
|
33
|
Ma L, Xu Y, Zhou J, Li Y, Zhang X, Jiang W, Wang G, Li R. Brain estrogen alters the effects of the antidepressant sertraline in middle-aged female and male mice. Mol Cell Endocrinol 2020; 516:110947. [PMID: 32702473 DOI: 10.1016/j.mce.2020.110947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/07/2020] [Accepted: 07/07/2020] [Indexed: 01/02/2023]
Abstract
Estrogens are important in regulating mood, especially for females. However, whether tissue-specific estrogen, such as brain estrogen, contributes to the effects of antidepressant treatment has not been determined. The present study used middle-aged aromatase gene knockout (Ar-/-) mice or overexpression (Thy1-Ar; hGFAP-Ar) mice as brain estrogen models to investigate whether brain estrogen synthesis alters the anti-depressive behaviors of sertraline treatment. Our results showed that depletion of brain estrogen increased depressive-like behavior in females, and elevated brain estrogen reduced depression-like behavior, regardless of sex. These genotype-related behaviors correlated with alterations of monoamine metabolism in the hippocampus (HPC) and the prefrontal cortex (PFC). We also demonstrated that male and female Ar-/- mice exhibited an attenuation of sertraline-induced anti-depressive behaviors compared to wild-type (WT) mice. The present data suggest that brain estrogen alters depressive-like behaviors and changes the effectiveness of antidepressants in middle-aged mice, regardless of sex.
Collapse
Affiliation(s)
- Lei Ma
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yong Xu
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China; Qingdao Municipal Hospital, Qingdao, China
| | - Jixuan Zhou
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yuhong Li
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Xinzhu Zhang
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Wei Jiang
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Gang Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| | - Rena Li
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
34
|
Zuloaga DG, Heck AL, De Guzman RM, Handa RJ. Roles for androgens in mediating the sex differences of neuroendocrine and behavioral stress responses. Biol Sex Differ 2020; 11:44. [PMID: 32727567 PMCID: PMC7388454 DOI: 10.1186/s13293-020-00319-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 07/09/2020] [Indexed: 12/17/2022] Open
Abstract
Estradiol and testosterone are powerful steroid hormones that impact brain function in numerous ways. During development, these hormones can act to program the adult brain in a male or female direction. During adulthood, gonadal steroid hormones can activate or inhibit brain regions to modulate adult functions. Sex differences in behavioral and neuroendocrine (i.e., hypothalamic pituitary adrenal (HPA) axis) responses to stress arise as a result of these organizational and activational actions. The sex differences that are present in the HPA and behavioral responses to stress are particularly important considering their role in maintaining homeostasis. Furthermore, dysregulation of these systems can underlie the sex biases in risk for complex, stress-related diseases that are found in humans. Although many studies have explored the role of estrogen and estrogen receptors in mediating sex differences in stress-related behaviors and HPA function, much less consideration has been given to the role of androgens. While circulating androgens can act by binding and activating androgen receptors, they can also act by metabolism to estrogenic molecules to impact estrogen signaling in the brain and periphery. This review focuses on androgens as an important hormone for modulating the HPA axis and behaviors throughout life and for setting up sex differences in key stress regulatory systems that could impact risk for disease in adulthood. In particular, impacts of androgens on neuropeptide systems known to play key roles in HPA and behavioral responses to stress (corticotropin-releasing factor, vasopressin, and oxytocin) are discussed. A greater knowledge of androgen action in the brain is key to understanding the neurobiology of stress in both sexes.
Collapse
Affiliation(s)
| | - Ashley L Heck
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | | | - Robert J Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
35
|
Giatti S, Diviccaro S, Falvo E, Garcia-Segura LM, Melcangi RC. Physiopathological role of the enzymatic complex 5α-reductase and 3α/β-hydroxysteroid oxidoreductase in the generation of progesterone and testosterone neuroactive metabolites. Front Neuroendocrinol 2020; 57:100836. [PMID: 32217094 DOI: 10.1016/j.yfrne.2020.100836] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/04/2020] [Accepted: 03/18/2020] [Indexed: 12/22/2022]
Abstract
The enzymatic complex 5α-reductase (5α-R) and 3α/3β-hydroxysteroid oxidoreductase (HSOR) is expressed in the nervous system, where it transforms progesterone (PROG) and testosterone (T) into neuroactive metabolites. These metabolites regulate myelination, brain maturation, neurotransmission, reproductive behavior and the stress response. The expression of 5α-R and 3α-HSOR and the levels of PROG and T reduced metabolites show regional and sex differences in the nervous system and are affected by changing physiological conditions as well as by neurodegenerative and psychiatric disorders. A decrease in their nervous tissue levels may negatively impact the course and outcome of some pathological events. However, in other pathological conditions their increased levels may have a negative impact. Thus, the use of synthetic analogues of these steroids or 5α-R modulation have been proposed as therapeutic approaches for several nervous system pathologies. However, further research is needed to fully understand the consequences of these manipulations, in particular with 5α-R inhibitors.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Eva Falvo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
36
|
Hamidovic A, Karapetyan K, Serdarevic F, Choi SH, Eisenlohr-Moul T, Pinna G. Higher Circulating Cortisol in the Follicular vs. Luteal Phase of the Menstrual Cycle: A Meta-Analysis. Front Endocrinol (Lausanne) 2020; 11:311. [PMID: 32582024 PMCID: PMC7280552 DOI: 10.3389/fendo.2020.00311] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/24/2020] [Indexed: 01/27/2023] Open
Abstract
Although results of animal research show that interactions between stress and sex hormones are implicated in the development of affective disorders in women, translation of these findings to patients has been scarce. As a basic step toward advancing this field of research, we analyzed findings of studies which reported circulating cortisol levels in healthy women in the follicular vs. luteal phase of the menstrual cycle. We deemed this analysis critical not only to advance our understanding of basic physiology, but also as an important contrast to the findings of future studies evaluating stress and sex hormones in women with affective disorders. We hypothesized that cortisol levels would be lower in the follicular phase based on the proposition that changes in levels of potent GABAergic neurosteroids, including allopregnanolone, during the menstrual cycle dynamically change in the opposite direction relative to cortisol levels. Implementing strict inclusion criteria, we compiled results of high-quality studies involving 778 study participants to derive a standardized mean difference between circulating cortisol levels in the follicular vs. luteal phase of the menstrual cycle. In line with our hypothesis, our meta-analysis found that women in the follicular phase had higher cortisol levels than women in the luteal phase, with an overall Hedges' g of 0.13 (p < 0.01) for the random effects model. No significant between-study difference was detected, with the level of heterogeneity in the small range. Furthermore, there was no evidence of publication bias. As cortisol regulation is a delicate process, we review some of the basic mechanisms by which progesterone, its potent metabolites, and estradiol regulate cortisol output and circulation to contribute to the net effect of higher cortisol in the follicular phase.
Collapse
Affiliation(s)
- Ajna Hamidovic
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, United States
- *Correspondence: Ajna Hamidovic
| | - Kristina Karapetyan
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, United States
| | - Fadila Serdarevic
- Department of Epidemiology, Erasmus Medical Centre Rotterdam, Rotterdam, Netherlands
| | - So Hee Choi
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, United States
| | - Tory Eisenlohr-Moul
- Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Graziano Pinna
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
37
|
Sze Y, Brunton PJ. Sex, stress and steroids. Eur J Neurosci 2019; 52:2487-2515. [DOI: 10.1111/ejn.14615] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Ying Sze
- Centre for Discovery Brain Sciences University of Edinburgh Edinburgh UK
| | - Paula J. Brunton
- Centre for Discovery Brain Sciences University of Edinburgh Edinburgh UK
- Zhejiang University‐University of Edinburgh Joint Institute Haining Zhejiang China
| |
Collapse
|
38
|
Duarte-Guterman P, Lieblich SE, Wainwright SR, Chow C, Chaiton JA, Watson NV, Galea LAM. Androgens Enhance Adult Hippocampal Neurogenesis in Males but Not Females in an Age-Dependent Manner. Endocrinology 2019; 160:2128-2136. [PMID: 31219567 PMCID: PMC6736050 DOI: 10.1210/en.2019-00114] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/24/2019] [Indexed: 01/27/2023]
Abstract
Androgens (testosterone and DHT) increase adult hippocampal neurogenesis by increasing survival of new neurons in male rats and mice via an androgen receptor pathway, but it is not known whether androgens regulate neurogenesis in female rats and whether the effect is age-dependent. We investigated the effects of DHT, a potent androgen, on neurogenesis in young adult and middle-aged male and female rats. Rats were gonadectomized and injected with the DNA synthesis marker bromodeoxyuridine (BrdU). The following day, rats began receiving daily injections of oil or DHT for 30 days. We evaluated cell proliferation (Ki67) and survival of new neurons (BrdU and BrdU/NeuN) in the hippocampus of male and female rats by using immunohistochemistry. As expected, DHT increased the number of BrdU+ cells in young males but surprisingly not in middle-aged males or in young and middle-aged females. In middle age, DHT increased the proportion of BrdU/NeuN cells, an effect driven by females. Androgen receptor expression also increased with aging in both female and male rats, which may contribute to a lack of DHT neurogenic effect in middle age. Our results indicate that DHT regulates adult hippocampal neurogenesis in a sex- and age-dependent manner.
Collapse
Affiliation(s)
- Paula Duarte-Guterman
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephanie E Lieblich
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Steven R Wainwright
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Carmen Chow
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jessica A Chaiton
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Neil V Watson
- Department of Psychology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Liisa A M Galea
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Correspondence: Liisa A. M. Galea, PhD, Djavad Mowafaghian Centre for Brain Health, 2215 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3, Canada. E-mail:
| |
Collapse
|
39
|
Heck AL, Handa RJ. Androgens Drive Sex Biases in Hypothalamic Corticotropin-Releasing Hormone Gene Expression After Adrenalectomy of Mice. Endocrinology 2019; 160:1757-1770. [PMID: 31074799 PMCID: PMC6594463 DOI: 10.1210/en.2019-00238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 05/05/2019] [Indexed: 12/16/2022]
Abstract
Although prominent sex differences exist in the hypothalamic-pituitary-adrenal axis's response to stressors, few studies of its regulation in the hypothalamic paraventricular nucleus (PVN) have compared both male and female subjects. In this study, we sought to explore sex differences in the acute regulation of PVN neuropeptide expression following glucocorticoid (GC) removal and the underlying role of gonadal hormones. We first examined the effects of short-term adrenalectomy (ADX) on PVN Crh and arginine vasopressin (Avp) expression in mice using in situ hybridization. ADX increased PVN AVP mRNA levels in both sexes. In contrast, PVN CRH mRNA was increased by 2 days after ADX in males only. Both sexes showed increases in CRH mRNA after 4 days. To determine if gonadal hormones contributed to this sex bias, we examined adrenalectomized (ADX'd) and gonadectomized (GDX'd) mice with or without gonadal hormone replacement. Unlike the pattern in intact animals, 2 days following ADX/gonadectomy, CRH mRNA levels did not increase in either sex. When males were given DHT propionate, CRH mRNA levels increased in ADX'd/GDX'd males similar to those observed following ADX alone. To determine a potential mechanism, we examined the coexpression of androgen receptor (AR) immunoreactivity and CRH neurons. Abundant colocalization was found in the anteroventral bed nucleus of the stria terminalis but not the PVN. Thus, our findings reveal a sex difference in PVN Crh expression following the removal of GC-negative feedback that may depend on indirect AR actions in males.
Collapse
Affiliation(s)
- Ashley L Heck
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Robert J Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
- Correspondence: Robert J. Handa, PhD, Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, Colorado 80523. E-mail:
| |
Collapse
|
40
|
Green MR, Zeidan M, Hodges TE, McCormick CM. Age-dependent regulation by androgens of gene expression in the anterior hypothalamus and stress-induced release of adrenal hormones in adolescent and adult male rats. J Neuroendocrinol 2019; 31:e12714. [PMID: 30912177 DOI: 10.1111/jne.12714] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 02/06/2019] [Accepted: 03/21/2019] [Indexed: 01/05/2023]
Abstract
Adolescents show greater and/or more prolonged activation of the hypothalamic-pituitary-adrenal axis in response to stressors than adults, although the basis for such an age difference is not understood. We investigated developmental shifts in the regulation of HPA function by testosterone using androgen replacement in orchiectomised (OCX) pre-pubertal and post-pubertal adolescent rats and in adults, as well as using inhibitors of testosterone synthesis in non-operated rats. The expected dampening effect of testosterone in adult OCX rats did not meet statistical significance in all of the three experiments. Nevertheless, in each, adolescents had higher post-stress concentrations of corticosterone compared to adults despite similar concentrations of testosterone. The effect of testosterone was in the opposite direction in post-pubertal adolescents compared to that in adults, with testosterone replacement leading to increased rather than lower corticosterone concentration. Testosterone replacement decreased arginine vasopressin and corticotrophin-releasing hormone immune-reactive cell counts in the paraventricular nucleus at all ages. In a fourth experiment, we provide evidence that the basis of the age difference in corticosterone release is because of a greater conversion of testosterone to oestradiol in adolescents and a greater conversion of testosterone to dihydrotestosterone in adults: aromatase inhibition had little effect in adults and attenuated the age difference by decreasing stress-induced corticosterone release in adolescents. By contrast, 5α-reductase inhibition or an androgen receptor antagonist had little effect in adolescents and attenuated the age difference by increasing stress-induced corticosterone release in adults. In the adrenal gland, adolescents had reduced 5α-reductase and androgen receptor gene expression. There also were age differences in the regulation of hypothalamic mRNA expression of androgen receptors, oestrogen receptors and aromatase by testosterone. In sum, the results suggest that developmental shifts in the synthesis of testosterone and the regulation of gene expression are factors with respect to age differences in corticosterone release in response to stressors.
Collapse
Affiliation(s)
- Matthew R Green
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
| | - Mostafa Zeidan
- Department of Biological Sciences, Brock University, St Catharines, Ontario, Canada
| | - Travis E Hodges
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
| | - Cheryl M McCormick
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
- Department of Biological Sciences, Brock University, St Catharines, Ontario, Canada
| |
Collapse
|
41
|
Effect of oestrogen-dependent vasopressin on HPA axis in the median eminence of female rats. Sci Rep 2019; 9:5153. [PMID: 30914732 PMCID: PMC6435644 DOI: 10.1038/s41598-019-41714-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/14/2019] [Indexed: 11/08/2022] Open
Abstract
The median eminence (ME) anatomically consists of external (eME) and internal (iME) layers. The hypothalamic neurosecretory cells terminate their axons in the eME and secrete their neurohormones regulating anterior pituitary hormone secretion involved in stress responses into the portal vein located in the eME. Magnocellular neurosecretory cells (MNCs) which produce arginine vasopressin (AVP) and oxytocin in the paraventricular (PVN) and supraoptic nuclei (SON) terminate their axons in the posterior pituitary gland (PP) through the iME. Here, we provide the first evidence that oestrogen modulates the dynamic changes in AVP levels in the eME axon terminals in female rats, using AVP-eGFP and AVP-DREADDs transgenic rats. Strong AVP-eGFP fluorescence in the eME was observed at all oestrus cycle stages in adult female rats but not in male transgenic rats. AVP-eGFP fluorescence in the eME was depleted after bilateral ovariectomy but re-appeared with high-dose 17β-oestradiol. AVP-eGFP fluorescence in the MNCs and PP did not change significantly in most treatments. Peripheral clozapine-N-oxide administration induced AVP-DREADDs neurone activation, causing a significant increase in plasma corticosterone levels in the transgenic rats. These results suggest that stress-induced activation of the hypothalamic-pituitary-adrenal axis may be caused by oestrogen-dependent upregulation of AVP in the eME of female rats.
Collapse
|
42
|
Herrera AY, Faude S, Nielsen SE, Locke M, Mather M. Effects of hormonal contraceptive phase and progestin generation on stress-induced cortisol and progesterone release. Neurobiol Stress 2019; 10:100151. [PMID: 30937356 PMCID: PMC6430619 DOI: 10.1016/j.ynstr.2019.100151] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/24/2019] [Accepted: 02/28/2019] [Indexed: 12/27/2022] Open
Abstract
The stress response differs between women using hormonal contraception and naturally cycling women. Yet, despite ample evidence showing that the stress response differs across the menstrual cycle in naturally cycling women, limited work has investigated whether the stress response differs across the hormonal contraceptive cycle, during which synthetic hormones are taken most of the month but not all of it. To induce a stress response, women using hormonal contraception completed the cold pressor test during either the active phase, when hormones are present, or during the inactive phase, when hormones are not present. Saliva was collected and assayed for free cortisol and progesterone levels prior to stress onset, immediately after stress termination, and 15-min post stress onset. Free cortisol and progesterone increased to a similar degree across both hormonal contraceptive phases in response to the cold pressor test. Post-hoc investigation indicates that the progestin “generation” (classification of synthetic progestins based on the compounds they are derived from) can differentially affect the free steroid response to cold pressor test stress, with the largest effects observed in women using formulations containing second-generation progestins. These findings indicate that progestin generation, particularly second-generation progestins, may have a more impactful influence on the stress response than hormonal contraceptive cycle phase. Potential mechanisms driving this effect and need for additional research are discussed.
Collapse
Affiliation(s)
- Alexandra Ycaza Herrera
- University of Southern California, Davis School of Gerontology, USA
- Corresponding author. University of Southern California, 3715 McClintock Ave, Rm 351, Los Angeles, 90089, California, USA.
| | | | - Shawn E. Nielsen
- University of Southern California, Davis School of Gerontology, USA
| | | | - Mara Mather
- University of Southern California, Davis School of Gerontology, USA
- University of Southern California, Department of Psychology, USA
- University of Southern California, Neuroscience Graduate Program, USA
| |
Collapse
|
43
|
Sex differences and the neurobiology of affective disorders. Neuropsychopharmacology 2019; 44:111-128. [PMID: 30061743 PMCID: PMC6235863 DOI: 10.1038/s41386-018-0148-z] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/14/2018] [Accepted: 06/25/2018] [Indexed: 12/11/2022]
Abstract
Observations of the disproportionate incidence of depression in women compared with men have long preceded the recent explosion of interest in sex differences. Nonetheless, the source and implications of this epidemiologic sex difference remain unclear, as does the practical significance of the multitude of sex differences that have been reported in brain structure and function. In this article, we attempt to provide a framework for thinking about how sex and reproductive hormones (particularly estradiol as an example) might contribute to affective illness. After briefly reviewing some observed sex differences in depression, we discuss how sex might alter brain function through hormonal effects (both organizational (programmed) and activational (acute)), sex chromosome effects, and the interaction of sex with the environment. We next review sex differences in the brain at the structural, cellular, and network levels. We then focus on how sex and reproductive hormones regulate systems implicated in the pathophysiology of depression, including neuroplasticity, genetic and neural networks, the stress axis, and immune function. Finally, we suggest several models that might explain a sex-dependent differential regulation of affect and susceptibility to affective illness. As a disclaimer, the studies cited in this review are not intended to be comprehensive but rather serve as examples of the multitude of levels at which sex and reproductive hormones regulate brain structure and function. As such and despite our current ignorance regarding both the ontogeny of affective illness and the impact of sex on that ontogeny, sex differences may provide a lens through which we may better view the mechanisms underlying affective regulation and dysfunction.
Collapse
|
44
|
Heck AL, Handa RJ. Sex differences in the hypothalamic-pituitary-adrenal axis' response to stress: an important role for gonadal hormones. Neuropsychopharmacology 2019; 44:45-58. [PMID: 30111811 PMCID: PMC6235871 DOI: 10.1038/s41386-018-0167-9] [Citation(s) in RCA: 276] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/19/2018] [Accepted: 07/22/2018] [Indexed: 12/11/2022]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis, a neuroendocrine network that controls hormonal responses to internal and external challenges in an organism's environment, exhibits strikingly sex-biased activity. In adult female rodents, acute HPA function following a stressor is markedly greater than it is in males, and this difference has largely been attributed to modulation by the gonadal hormones testosterone and estradiol. These gonadal hormones are produced by the hypothalamic-pituitary-gonadal (HPG) axis and have been shown to determine sex differences in adult HPA function after acute stress via their activational and organizational effects. Although these actions of gonadal hormones are well supported, the possibility that sex chromosomes similarly influence HPA activity is unexplored. Moreover, questions remain regarding sex differences in the activity of the HPA axis following chronic stress and the underlying contributions of gonadal hormones and sex chromosomes. The present review examines what is currently known about sex differences in the neuroendocrine response to stress, as well as outstanding questions regarding this sex bias. Although it primarily focuses on the rodent literature, a brief discussion of sex differences in the human HPA axis is also included.
Collapse
Affiliation(s)
- Ashley L. Heck
- 0000 0004 1936 8083grid.47894.36Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523 USA
| | - Robert J. Handa
- 0000 0004 1936 8083grid.47894.36Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523 USA
| |
Collapse
|
45
|
Rosinger ZJ, Jacobskind JS, Bulanchuk N, Malone M, Fico D, Justice NJ, Zuloaga DG. Characterization and gonadal hormone regulation of a sexually dimorphic corticotropin-releasing factor receptor 1 cell group. J Comp Neurol 2018; 527:1056-1069. [PMID: 30499109 DOI: 10.1002/cne.24588] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/16/2018] [Accepted: 11/09/2018] [Indexed: 12/11/2022]
Abstract
Corticotropin-releasing factor binds with high affinity to CRF receptor 1 (CRFR1) and is implicated in stress-related mood disorders such as anxiety and depression. Using a validated CRFR1-green fluorescent protein (GFP) reporter mouse, our laboratory recently discovered a nucleus of CRFR1 expressing cells that is prominent in the female rostral anteroventral periventricular nucleus (AVPV/PeN), but largely absent in males. This sex difference is present in the early postnatal period and remains dimorphic into adulthood. The present investigation sought to characterize the chemical composition and gonadal hormone regulation of these sexually dimorphic CRFR1 cells using immunohistochemical procedures. We report that CRFR1-GFP-ir cells within the female AVPV/PeN are largely distinct from other dimorphic cell populations (kisspeptin, tyrosine hydroxylase). However, CRFR1-GFP-ir cells within the AVPV/PeN highly co-express estrogen receptor alpha as well as glucocorticoid receptor. A single injection of testosterone propionate or estradiol benzoate on the day of birth completely eliminates the AVPV/PeN sex difference, whereas adult gonadectomy has no effect on CRFR1-GFP cell number. These results indicate that the AVPV/PeN CRFR1 is regulated by perinatal but not adult gonadal hormones. Finally, female AVPV/PeN CRFR1-GFP-ir cells are activated following an acute 30-min restraint stress, as assessed by co-localization of CRFR1-GFP cells with phosphorylated (p) CREB. CRFR1-GFP/pCREB cells were largely absent in the male AVPV/PeN. Together, these data indicate a stress and gonadal hormone responsive nucleus that is unique to females and may contribute to sex-specific stress responses.
Collapse
Affiliation(s)
| | | | - Nicole Bulanchuk
- Department of Psychology, University at Albany, Albany, New York
| | - Margaret Malone
- Department of Psychology, University at Albany, Albany, New York
| | - Danielle Fico
- Department of Psychology, University at Albany, Albany, New York
| | - Nicholas J Justice
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Sciences Center, Houston, Texas
| | - Damian G Zuloaga
- Department of Psychology, University at Albany, Albany, New York
| |
Collapse
|
46
|
Acevedo-Rodriguez A, Kauffman AS, Cherrington BD, Borges CS, Roepke TA, Laconi M. Emerging insights into hypothalamic-pituitary-gonadal axis regulation and interaction with stress signalling. J Neuroendocrinol 2018; 30. [PMID: 29524268 PMCID: PMC6129417 DOI: 10.1111/jne.12590] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Reproduction and fertility are regulated via hormones of the hypothalamic-pituitary-gonadal (HPG) axis. Control of this reproductive axis occurs at all levels, including the brain and pituitary, and allows for the promotion or inhibition of gonadal sex steroid secretion and function. In addition to guiding proper gonadal development and function, gonadal sex steroids also act in negative- and positive-feedback loops to regulate reproductive circuitry in the brain, including kisspeptin neurones, thereby modulating overall HPG axis status. Additional regulation is also provided by sex steroids made within the brain, including neuroprogestins. Furthermore, because reproduction and survival need to be coordinated and balanced, the HPG axis is able to modulate (and be modulated by) stress hormone signalling, including cortiscosterone, from the hypothalamic-pituitary-adrenal (HPA) axis. This review covers recent data related to the neural, hormonal and stress regulation of the HPG axis and emerging interactions between the HPG and HPA axes, focusing on actions at the level of the brain and pituitary.
Collapse
Affiliation(s)
- A Acevedo-Rodriguez
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - A S Kauffman
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - B D Cherrington
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA
| | - C S Borges
- Department of Morphology, Institute of Biosciences, São Paulo State University (Unesp), Botucatu, Brazil
| | - T A Roepke
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - M Laconi
- Laboratorio de Fisiopatología Ovárica, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU - CONICET), Universidad Juan Agustín Maza, Mendoza, Argentina
- Facultad de Ciencias Veterinarias y Ambientales, Universidad Juan Agustín Maza, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad de Mendoza, Mendoza, Argentina
| |
Collapse
|
47
|
Zhang R, Zhang Y, Wu M, Yan P, Izaz A, Wang R, Zhu H, Zhou Y, Wu X. Molecular cloning of androgen receptor and gene expression of sex steroid hormone receptors in the brain of newborn Chinese alligator (Alligator sinensis). Gene 2018; 674:178-187. [DOI: 10.1016/j.gene.2018.06.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 05/29/2018] [Accepted: 06/11/2018] [Indexed: 12/16/2022]
|
48
|
Abstract
Accumulating research indicates oxytocin plays a significant role in regulating the behavioral and neurobiological responses to stress. Evidence from preclinical models suggests the effect of oxytocin on stress-responsivity appears to be dependent on individual characteristics, including sex. Although the interaction between oxytocinergic and stress systems has been widely studied in rodents, recent efforts have been made to examine the interface between these two systems in humans. This brief review examines how administration of oxytocin can influence the neuroendocrine, behavioral, and neural responses to stress, explores how sex may impact these effects, and provides considerations for future work.
Collapse
|
49
|
Prepubertal Development of GABAergic Transmission to Gonadotropin-Releasing Hormone (GnRH) Neurons and Postsynaptic Response Are Altered by Prenatal Androgenization. J Neurosci 2018; 38:2283-2293. [PMID: 29374136 DOI: 10.1523/jneurosci.2304-17.2018] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 12/29/2017] [Accepted: 01/20/2018] [Indexed: 11/21/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) neurons regulate reproduction through pulsatile GnRH release. Women with polycystic ovary syndrome (PCOS) have persistently elevated luteinizing hormone release frequency, reflecting GnRH release; this exacerbates hyperandrogenemia and disrupted reproductive cycles that are characteristic of this disorder. Clinical evidence suggests that neuroendocrine features of PCOS may manifest peripubertally. Adult mice prenatally exposed to androgens (PNA) mimic several reproductive features of PCOS. GnRH neurons from these mice have increased firing activity and receive increased GABAergic transmission, which is excitatory. When changes emerge during development is unknown. To study the typical postnatal development of GABAergic transmission and the effects of PNA treatment and sex, whole-cell voltage-clamp recordings were made of GABAergic postsynaptic currents (PSCs) in GnRH neurons in brain slices from prepubertal through adult control and PNA female and male mice. GABAergic transmission was present by 1 week of age in females and males and increased in frequency, reaching adult levels at 3 and 4 weeks, respectively. GABAergic PSC frequency was elevated in 3-week-old PNA versus control females. PSC frequency in both controls and PNA mice was activity independent, suggesting that PNA induces changes in synapse organization. PNA also alters the functional response of GnRH neurons to GABA. GABA induced firing in fewer neurons from 3-week-old PNA than control females; membrane potential depolarization induced by GABA was also reduced in cells from PNA mice at this age. PNA thus induces changes during development in the presynaptic organization of the GABAergic network afferent to GnRH neurons as well as the postsynaptic GnRH neuron response, both of which may contribute to adult reproductive dysfunction.SIGNIFICANCE STATEMENT The central neuronal network that regulates reproduction is overactive in polycystic ovary syndrome (PCOS), a leading cause of infertility. Recent evidence of neuroendocrine dysfunction in midpubertal girls suggests that the pathophysiological mechanisms underlying PCOS may arise before pubertal maturation. Prenatal exposure to androgens (PNA) in mice mimics several neuroendocrine features of PCOS. GABAergic transmission to gonadotropin-releasing hormone (GnRH) neurons is important for reproduction and is increased in adult PNA mice. The typical development of this network and when changes with PNA and sex arise relative to puberty are unknown. These studies provide evidence that PNA alters prepubertal development of the GABAergic network afferent to GnRH neurons, including both the presynaptic organization and postsynaptic response. These changes may contribute to reproductive dysfunction in adults.
Collapse
|
50
|
Abstract
Biosynthesis and secretion of the hypothalamic nonapeptide oxytocin largely depends on steroid hormones. Estradiol, corticosterone, and vitamin D seem to be the most prominent actors. Due to their lipophilic nature, systemic steroids are thought to be capable of crossing the blood-brain barrier, thus mediating central functions including neuroendocrine and behavioral control. The actual mode of action of steroids in hypothalamic circuitry is still unknown: Most of the oxytocinergic perikarya lack nuclear steroid receptors but express proteins suspected to be membrane receptors for steroids. Oxytocin expressing neurons contain enzymes important for intrinsic steroid metabolism. Furthermore, they produce and probably liberate specific steroid-binding globulins. Rapid responses to steroid hormones may involve these binding proteins and membrane-associated receptors, rather than classic nuclear receptors and genomic pathways. Neuroendocrine regulation, reproductive behaviors, and stress response seem to depend on these mechanisms.
Collapse
Affiliation(s)
| | - Scott D Ochs
- Dept. of Pharmacology, Via College of Osteopathic Medicine, Spartanburg, SC, USA
| | - Jack D Caldwell
- Dept. of Pharmacology, Via College of Osteopathic Medicine, Spartanburg, SC, USA
| |
Collapse
|