1
|
Cárdenas-Bedoya J, Torres-Mendoza BM, Martínez-Torres NI. Cerebrolysin Induces Dendritic Tree Plastic Changes and BDNF Increase in the Amygdala of Male Rats with Maternal Deprivation. Neurochem Res 2025; 50:123. [PMID: 40123021 DOI: 10.1007/s11064-025-04376-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Several psychopathologies may be triggered, among other factors, by stress or trauma in childhood. The maternal deprivation model (MD) replicates some of the core factors that may induce the onset of different psychopathologies like structural and plasticity defects. Among other brain regions, the amygdala is susceptible to stress and trauma and plays a key role in integrating social behavior. Several molecules, such as Cerebrolysin® (CBL), have been used to treat different symptoms by reversing the underlying neurobiological plasticity-related changes. In this study, maternal deprivation (MD) was conducted on 9-day-old male Sprague-Dawley rats (n = 28; 7 rats per group: Intact, Intact + CBL, MD, and MD + CBL). At 25 postnatal days (PND), CBL treatment was administered for 10 consecutive days, and social behavior was evaluated in a three-chamber social test at 35 PND. Moreover, Sholl analysis and immunohistochemistry were carried out for dendritic intersection and brain-derived neurotrophic factor (BDNF) presence in the amygdala, respectively. CBL treatment had an augmentative effect on intact animals in terms of social behavior and incidences, but no differences between MD and CBL-treated animals. Moreover, dendritic intersections and BDNF decreased after the MD protocol but increased by CBL treatment in MD animals. Our results show that CBL could be part of the treatment in case of a traumatic or stressful event in neurodevelopment, especially in the youth age. According to this preclinical study, CBL could help reverse symptoms of different psychopathologies caused by stress or trauma, with neurobiological changes underlying its effect.
Collapse
Affiliation(s)
- Jhonathan Cárdenas-Bedoya
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Col. Independencia, Guadalajara, C.P. 44340, Jalisco, México
- Centro Universitario de Ciencias de la Salud, Departamento de Disciplinas Filósofico, Metodológicas e Instrumentales, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Blanca Miriam Torres-Mendoza
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Col. Independencia, Guadalajara, C.P. 44340, Jalisco, México
- Centro Universitario de Ciencias de la Salud, Departamento de Disciplinas Filósofico, Metodológicas e Instrumentales, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Nestor Ismael Martínez-Torres
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Col. Independencia, Guadalajara, C.P. 44340, Jalisco, México.
- Departamento de Bienestar y Desarrollo Sustentable, Centro Universitario del Norte, Universidad de Guadalajara, Colotlán, Jalisco, México.
| |
Collapse
|
2
|
Kaplan HS, Horvath PM, Rahman MM, Dulac C. The neurobiology of parenting and infant-evoked aggression. Physiol Rev 2025; 105:315-381. [PMID: 39146250 DOI: 10.1152/physrev.00036.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 07/19/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Parenting behavior comprises a variety of adult-infant and adult-adult interactions across multiple timescales. The state transition from nonparent to parent requires an extensive reorganization of individual priorities and physiology and is facilitated by combinatorial hormone action on specific cell types that are integrated throughout interconnected and brainwide neuronal circuits. In this review, we take a comprehensive approach to integrate historical and current literature on each of these topics across multiple species, with a focus on rodents. New and emerging molecular, circuit-based, and computational technologies have recently been used to address outstanding gaps in our current framework of knowledge on infant-directed behavior. This work is raising fundamental questions about the interplay between instinctive and learned components of parenting and the mutual regulation of affiliative versus agonistic infant-directed behaviors in health and disease. Whenever possible, we point to how these technologies have helped gain novel insights and opened new avenues of research into the neurobiology of parenting. We hope this review will serve as an introduction for those new to the field, a comprehensive resource for those already studying parenting, and a guidepost for designing future studies.
Collapse
Affiliation(s)
- Harris S Kaplan
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Patricia M Horvath
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Mohammed Mostafizur Rahman
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Catherine Dulac
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| |
Collapse
|
3
|
Bencker C, Gschwandtner L, Nayman S, Grikšienė R, Nguyen B, Nater UM, Guennoun R, Sundström-Poromaa I, Pletzer B, Bixo M, Comasco E. Progestagens and progesterone receptor modulation: Effects on the brain, mood, stress, and cognition in females. Front Neuroendocrinol 2025; 76:101160. [PMID: 39515587 DOI: 10.1016/j.yfrne.2024.101160] [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: 08/19/2024] [Revised: 10/28/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Progesterone is a highly lipophilic gonadal hormone that can influence behavior and mental health through its receptors in the brain. Fluctuations in progesterone levels across critical periods of a females life are associated with increased susceptibility to mental conditions. This review highlights the effects of progestagens, including progesterone and synthetic progestins, on the brain, mood, stress, and cognition in females. The primary focus is on experimental pharmacological research that teases out the distinct effects of progestagens from those of estrogens. Additionally, the key literature on puberty, the menstrual cycle, pregnancy, perimenopause, hormonal contraceptives, and menopausal hormone therapy is reviewed, although conclusions are limited by the nested effects of progestagens and estrogens. Single study-findings suggest an influence of progesterone on amygdala reactivity related to processing of emotional stimuli and memory. In patients with premenstrual dysphoric disorder, progesterone receptor modulation improves premenstrual mood symptoms and potentially enhances fronto-cingulate control over emotion processing. The interaction between progestagens and the systems involved in the regulation of stress seems to influence subjective experiences of mood and stress. Sparse studies investigating the effects of progestin-only contraceptives suggest effects of progestagens on the brain, mood, and stress. Progesterone and progestins used for contraception can influence neural processes as myelination and neuroprotection, exerting protective effects against stroke. Concerning menopausal hormonal therapy, the effects of progestins are largely unknown. Levels of progesterone as well as type, administration route, timing, dose regimen, metabolism, and intracellular activity of progestins in hormonal contraceptives and menopausal hormonal therapy are factors whose effects remain to be elucidated. Altogether, current knowledge highlights the potential role of progestagens in females health but also calls for well-designed pharmaco-behavioral studies disentangling the effects of progestagens from those of estrogens.
Collapse
Affiliation(s)
- Celine Bencker
- Department of Clinical and Health Psychology, Faculty of Psychology, University of Vienna, Vienna, Austria; University Research Platform "Stress of Life (SOLE) - Processes and Mechanisms underlying Everyday Life Stress", University of Vienna, Vienna, Austria
| | - Laura Gschwandtner
- Department of Clinical and Health Psychology, Faculty of Psychology, University of Vienna, Vienna, Austria; University Research Platform "Stress of Life (SOLE) - Processes and Mechanisms underlying Everyday Life Stress", University of Vienna, Vienna, Austria
| | - Sibel Nayman
- Research Group Longitudinal and Intervention Research, Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, Mannheim, Germany
| | - Ramunė Grikšienė
- Department of Neurobiology and Biophysics, Life Science Center, Vilnius University, Lithuania
| | | | - Urs M Nater
- Department of Clinical and Health Psychology, Faculty of Psychology, University of Vienna, Vienna, Austria; University Research Platform "Stress of Life (SOLE) - Processes and Mechanisms underlying Everyday Life Stress", University of Vienna, Vienna, Austria
| | | | | | - Belinda Pletzer
- Department of Psychology, Centre for Cognitive Neuroscience, University of Salzburg, Austria
| | - Marie Bixo
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, Sweden
| | - Erika Comasco
- Department of Women's and Children's Health, Science for Life Laboratory, Uppsala University, Sweden.
| |
Collapse
|
4
|
Cioffi L, Diviccaro S, Chrostek G, Caruso D, Garcia-Segura LM, Melcangi RC, Giatti S. Neuroactive steroids fluctuate with regional specificity in the central and peripheral nervous system across the rat estrous cycle. J Steroid Biochem Mol Biol 2024; 243:106590. [PMID: 39053702 DOI: 10.1016/j.jsbmb.2024.106590] [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: 06/25/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Neuroactive steroids (i.e., sex steroid hormones and neurosteroids) are important physiological regulators of nervous function and potential neuroprotective agents for neurodegenerative and psychiatric disorders. Sex is an important component of such effects. However, even if fluctuations in sex steroid hormone level during the menstrual cycle are associated with neuropathological events in some women, the neuroactive steroid pattern in the brain across the ovarian cycle has been poorly explored. Therefore, we assessed the levels of pregnenolone, progesterone, and its metabolites (i.e., dihydroprogesterone, allopregnanolone and isoallopregnanolone), dehydroepiandrosterone, testosterone and its metabolites (i.e., dihydrotestosterone, 3α-diol and 17β-estradiol) across the rat ovarian cycle to determine whether their plasma fluctuations are similar to those occurring in the central (i.e., hippocampus and cerebral cortex) and peripheral (i.e., sciatic nerve) nervous system. Data obtained indicate that the plasma pattern of these molecules generally does not fully reflect the events occurring in the nervous system. In addition, for some neuroactive steroid levels, the pattern is not identical between the two brain regions and between the brain and peripheral nerves. Indeed, with the exception of progesterone, all other neuroactive steroids assessed here showed peculiar regional differences in their pattern of fluctuation in the nervous system during the estrous cycle. These observations may have important diagnostic and therapeutic consequences for neuropathological events influenced by the menstrual cycle.
Collapse
Affiliation(s)
- Lucia Cioffi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| | - Silvia Diviccaro
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| | - Gabriela Chrostek
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| | - Luis Miguel Garcia-Segura
- Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain and Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy.
| | - Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Italy
| |
Collapse
|
5
|
Sgobbi RF, Incrocci RM, Paliarin F, Nobre MJ. The modulatory role of serotonin-1A receptors of the basolateral amygdala and dorsal periaqueductal gray on the impact of hormonal variation on the conditioned fear response. Neuroscience 2024; 554:118-127. [PMID: 39019393 DOI: 10.1016/j.neuroscience.2024.07.017] [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: 02/23/2024] [Revised: 06/07/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
Despite significant advances in the study of fear and fear memory formation, little is known about fear learning and expression in females. This omission has been proven surprising, as normal and pathological behaviors are highly influenced by ovarian hormones, particularly estradiol and progesterone. In the current study, we investigated the joint influence of serotonin (5-HT) neurotransmission and estrous cycle phases (low or high levels of estradiol and progesterone) on the expression of conditioned fear in a group of female rats that were previously divided according to their response to stressful stimuli into low or high anxiety-like subjects. The baseline amplitude of the unconditioned acoustic startle responses was high in high-anxiety female rats, with no effect on the estrous cycle observed. Data collected during the proestrus-estrus phase revealed that low-anxiety rats had startle amplitudes similar to those of high-anxiety rats. It is supposed that high-anxiety female rats benefit from increased estradiol and progesterone levels to achieve comparable potentiated startle amplitudes. In contrast, female rats experienced a significant decrease in hormone levels during the Diestrus phase. This decrease is believed to play a role in preventing them from displaying a heightened startle response when faced with strongly aversive stimuli. Data collected after 5-HT and 8-OH-DPAT were administered into the basolateral nuclei and dorsal periaqueductal gray suggest that 5-HT neurotransmission works with progesterone and estrogen to reduce startle potentiation, most likely by activating the serotonin-1A receptor subtype.
Collapse
Affiliation(s)
- R F Sgobbi
- Departamento de Psicologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo (USP), 14040-901 Ribeirão Preto, SP, Brasil
| | - R M Incrocci
- Departamento de Psicologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo (USP), 14040-901 Ribeirão Preto, SP, Brasil
| | - F Paliarin
- Departamento de Psicologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo (USP), 14040-901 Ribeirão Preto, SP, Brasil
| | - M J Nobre
- Departamento de Psicologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo (USP), 14040-901 Ribeirão Preto, SP, Brasil; Departamento de Psicologia, Uni-FACEF, 14401-135, Franca, SP, Brasil.
| |
Collapse
|
6
|
He X, Hou F, Shen X, Zhao D, Zhao X, Qi M, Li P. Individual and additive-effect relationships of menopausal symptoms and subjective cognitive decline among nurses during menopausal transition: a cross-sectional study. Menopause 2024; 31:617-625. [PMID: 38860940 DOI: 10.1097/gme.0000000000002368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
OBJECTIVE This study aimed to examine the individual and additive-effect relationships between menopausal symptoms and subjective cognitive decline among nurses during menopausal transition. METHODS Between February and September 2019, a convenience sampling strategy was used, involving 1,335 Chinese nurses undergoing menopausal transition. A general information survey that included the Subjective Cognitive Decline Scale and the Menopause Rating Scale was completed. Based on a cut-off point of the subjective cognitive decline score of 7.5, the overall sample was divided into mild and severe groups. Propensity score matching was performed to balance covariates of mild and severe subjective cognitive decline. The individual and cumulative effects of menopausal symptoms and subjective cognitive decline were analyzed using binary logistic regression and the Cochran-Armitage trend test, respectively. RESULTS After propensity score matching, none of the parameters showed significant differences between the groups. Logistic regression analysis revealed that four menopausal symptoms were closely associated with severe subjective cognitive decline. The Cochran-Armitage trend test indicated odds ratios linking the presence of these symptoms with increased severe subjective cognitive impairment. In addition, nurses simultaneously experiencing two or more core menopausal symptoms were over six times more likely to have severe subjective cognitive decline than nurses experiencing none or one core menopausal symptom during menopausal transition. CONCLUSIONS Individual and additive numbers of menopausal symptoms significantly influenced subjective cognitive decline in nurses during their menopausal transition. These findings suggest that interventions aimed at enhancing the cognitive performance of nurses experiencing menopause should consider menopausal symptoms.
Collapse
Affiliation(s)
- Xudong He
- From the School of Nursing and Rehabilitation, Shandong University, Jinan, Shandong, China
| | - Fangyan Hou
- Shandong Youth Political College, Jinan, Shandong, China
| | - Xiaona Shen
- From the School of Nursing and Rehabilitation, Shandong University, Jinan, Shandong, China
| | - Di Zhao
- From the School of Nursing and Rehabilitation, Shandong University, Jinan, Shandong, China
| | - Xiangyu Zhao
- From the School of Nursing and Rehabilitation, Shandong University, Jinan, Shandong, China
| | - Meiling Qi
- From the School of Nursing and Rehabilitation, Shandong University, Jinan, Shandong, China
| | - Ping Li
- From the School of Nursing and Rehabilitation, Shandong University, Jinan, Shandong, China
| |
Collapse
|
7
|
Singh M, Krishnamoorthy VR, Kim S, Khurana S, LaPorte HM. Brain-derived neuerotrophic factor and related mechanisms that mediate and influence progesterone-induced neuroprotection. Front Endocrinol (Lausanne) 2024; 15:1286066. [PMID: 38469139 PMCID: PMC10925611 DOI: 10.3389/fendo.2024.1286066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/15/2024] [Indexed: 03/13/2024] Open
Abstract
Historically, progesterone has been studied significantly within the context of reproductive biology. However, there is now an abundance of evidence for its role in regions of the central nervous system (CNS) associated with such non-reproductive functions that include cognition and affect. Here, we describe mechanisms of progesterone action that support its brain-protective effects, and focus particularly on the role of neurotrophins (such as brain-derived neurotrophic factor, BDNF), the receptors that are critical for their regulation, and the role of certain microRNA in influencing the brain-protective effects of progesterone. In addition, we describe evidence to support the particular importance of glia in mediating the neuroprotective effects of progesterone. Through this review of these mechanisms and our own prior published work, we offer insight into why the effects of a progestin on brain protection may be dependent on the type of progestin (e.g., progesterone versus the synthetic, medroxyprogesterone acetate) used, and age, and as such, we offer insight into the future clinical implication of progesterone treatment for such disorders that include Alzheimer's disease, stroke, and traumatic brain injury.
Collapse
Affiliation(s)
- Meharvan Singh
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | | | | | | | | |
Collapse
|
8
|
Arjmand S, Bender D, Jakobsen S, Wegener G, Landau AM. Peering into the Brain's Estrogen Receptors: PET Tracers for Visualization of Nuclear and Extranuclear Estrogen Receptors in Brain Disorders. Biomolecules 2023; 13:1405. [PMID: 37759805 PMCID: PMC10526964 DOI: 10.3390/biom13091405] [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: 08/10/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Estrogen receptors (ERs) play a multitude of roles in brain function and are implicated in various brain disorders. The use of positron emission tomography (PET) tracers for the visualization of ERs' intricate landscape has shown promise in oncology but remains limited in the context of brain disorders. Despite recent progress in the identification and development of more selective ligands for various ERs subtypes, further optimization is necessary to enable the reliable and efficient imaging of these receptors. In this perspective, we briefly touch upon the significance of estrogen signaling in the brain and raise the setbacks associated with the development of PET tracers for identification of specific ERs subtypes in the brain. We then propose avenues for developing efficient PET tracers to non-invasively study the dynamics of ERs in the brain, as well as neuropsychiatric diseases associated with their malfunction in a longitudinal manner. This perspective puts several potential candidates on the table and highlights the unmet needs and areas requiring further research to unlock the full potential of PET tracers for ERs imaging, ultimately aiding in deepening our understanding of ERs and forging new avenues for potential therapeutic strategies.
Collapse
Affiliation(s)
- Shokouh Arjmand
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| | - Dirk Bender
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| | - Steen Jakobsen
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
| | - Anne M. Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| |
Collapse
|
9
|
Gundacker A, Cuenca Rico L, Stoehrmann P, Tillmann KE, Weber-Stadlbauer U, Pollak DD. Interaction of the pre- and postnatal environment in the maternal immune activation model. DISCOVER MENTAL HEALTH 2023; 3:15. [PMID: 37622027 PMCID: PMC10444676 DOI: 10.1007/s44192-023-00042-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023]
Abstract
Adverse influences during pregnancy are associated with a range of unfavorable outcomes for the developing offspring. Maternal psychosocial stress, exposure to infections and nutritional imbalances are known risk factors for neurodevelopmental derangements and according psychiatric and neurological manifestations later in offspring life. In this context, the maternal immune activation (MIA) model has been extensively used in preclinical research to study how stimulation of the maternal immune system during gestation derails the tightly coordinated sequence of fetal neurodevelopment. The ensuing consequence of MIA for offspring brain structure and function are majorly manifested in behavioral and cognitive abnormalities, phenotypically presenting during the periods of adolescence and adulthood. These observations have been interpreted within the framework of the "double-hit-hypothesis" suggesting that an elevated risk for neurodevelopmental disorders results from an individual being subjected to two adverse environmental influences at distinct periods of life, jointly leading to the emergence of pathology. The early postnatal period, during which the caregiving parent is the major determinant of the newborn´s environment, constitutes a window of vulnerability to external stimuli. Considering that MIA not only affects the developing fetus, but also impinges on the mother´s brain, which is in a state of heightened malleability during pregnancy, the impact of MIA on maternal brain function and behavior postpartum may importantly contribute to the detrimental consequences for her progeny. Here we review current information on the interaction between the prenatal and postnatal maternal environments in the modulation of offspring development and their relevance for the pathophysiology of the MIA model.
Collapse
Affiliation(s)
- Anna Gundacker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| | - Laura Cuenca Rico
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| | - Peter Stoehrmann
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| | - Katharina E. Tillmann
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Daniela D. Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| |
Collapse
|
10
|
Pletzer B, Winkler-Crepaz K, Hillerer K. Progesterone and contraceptive progestin actions on the brain: A systematic review of animal studies and comparison to human neuroimaging studies. Front Neuroendocrinol 2023; 69:101060. [PMID: 36758768 DOI: 10.1016/j.yfrne.2023.101060] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 01/25/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
In this review we systematically summarize the effects of progesterone and synthetic progestins on neurogenesis, synaptogenesis, myelination and six neurotransmitter systems. Several parallels between progesterone and older generation progestin actions emerged, suggesting actions via progesterone receptors. However, existing results suggest a general lack of knowledge regarding the effects of currently used progestins in hormonal contraception regarding these cellular and molecular brain parameters. Human neuroimaging studies were reviewed with a focus on randomized placebo-controlled trials and cross-sectional studies controlling for progestin type. The prefrontal cortex, amygdala, salience network and hippocampus were identified as regions of interest for future preclinical studies. This review proposes a series of experiments to elucidate the cellular and molecular actions of contraceptive progestins in these areas and link these actions to behavioral markers of emotional and cognitive functioning. Emotional effects of contraceptive progestins appear to be related to 1) alterations in the serotonergic system, 2) direct/indirect modulations of inhibitory GABA-ergic signalling via effects on the allopregnanolone content of the brain, which differ between androgenic and anti-androgenic progestins. Cognitive effects of combined oral contraceptives appear to depend on the ethinylestradiol dose.
Collapse
Affiliation(s)
- Belinda Pletzer
- Department of Psychology & Centre for Cognitive Neuroscience, Paris-Lodron-University Salzburg, Salzburg Austria.
| | | | - Katharina Hillerer
- Department of Gynaecology & Obstetrics, Private Medical University, Salzburg, Austria
| |
Collapse
|
11
|
Hampson E. Oral contraceptives in the central nervous system: Basic pharmacology, methodological considerations, and current state of the field. Front Neuroendocrinol 2023; 68:101040. [PMID: 36243109 DOI: 10.1016/j.yfrne.2022.101040] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/19/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
Millions of women around the world use combined oral contraceptives (OCs), yet surprisingly little is known about their central nervous system (CNS) effects. This article provides a short overview of the basic pharmacology of OCs, emphasizing features that may be relevant to understanding their effects in the CNS. Historical and recent findings from studies of cognitive function, mood, and negative affect (depressive changes under OC use) are then reviewed. We also present data from an archival dataset from our own laboratory in which we explore dysphoric changes in women using four generations of contraceptive progestins. Current data in the field are consistent with a modest effect of OC use on CNS variables, but conclusions based on current findings must be made very cautiously because of multiple methodological issues in many published studies to date, and inconsistencies in the findings. Directions for future research over the next 10 years are suggested. (150 words).
Collapse
Affiliation(s)
- Elizabeth Hampson
- Department of Psychology, University of Western Ontario, London, ON, Canada; Department of Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
12
|
Grant AD, Erickson EN. Birth, love, and fear: Physiological networks from pregnancy to parenthood. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2022; 11:100138. [PMID: 35757173 PMCID: PMC9227990 DOI: 10.1016/j.cpnec.2022.100138] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 12/01/2022] Open
Abstract
Pregnancy and childbirth are among the most dramatic physiological and emotional transformations of a lifetime. Despite their central importance to human survival, many gaps remain in our understanding of the temporal progression of and mechanisms underlying the transition to new parenthood. The goal of this paper is to outline the physiological and emotional development of the maternal-infant dyad from late pregnancy to the postpartum period, and to provide a framework to investigate this development using non-invasive timeseries. We focus on the interaction among neuroendocrine, emotional, and autonomic outputs in the context of late pregnancy, parturition, and post-partum. We then propose that coupled dynamics in these outputs can be leveraged to map both physiologic and pathologic pregnancy, parturition, and parenthood. This approach could address gaps in our knowledge and enable early detection or prediction of problems, with both personalized depth and broad population scale.
Collapse
Affiliation(s)
- Azure D. Grant
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, United States
- Levels Health Inc., 228 Park Ave. South, PMB 63877, New York, NY, 10003, United States
| | - Elise N. Erickson
- Oregon Health and Science University, Portland, OR, 97239, United States
| |
Collapse
|
13
|
Kaur M, Kaur M. Cognitive performance of women at various stages of reproductive aging and associated risk factors. Climacteric 2022; 25:278-285. [PMID: 34382492 DOI: 10.1080/13697137.2021.1956452] [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: 06/02/2021] [Revised: 07/05/2021] [Accepted: 07/12/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Menopausal transition among midlife women is accompanied by the issues of hot flushes, mood swings as well as sleep disturbances, night sweats, urogenital diminution and cognitive changes. The present cross-sectional study is an attempt to recognize the cognitive differences associated with various stages of reproductive aging. METHODS The study included women (N = 404) aged between 40 and 65 years from rural areas of Punjab (North India). The Mini-Mental State Examination (Hindi version) was used to evaluate orientation, registration, attention, recall, language and visual spatial skills domains of the global cognitive functioning of all participants. RESULTS The results of the present study exhibited a decline in cognitive scores across successive menopausal transitional stages in most of the cognitive domains except registration. The results of multivariate regression analysis demonstrated that illiteracy, vegetarian diet, perimenopause and late postmenopause phase were significantly associated with lower global cognitive scores. CONCLUSION With increased longevity, early identification of potential risk factors associated with cognitive decline among women during their midlife can be beneficial in improving the mental health of postmenopausal women.
Collapse
Affiliation(s)
- M Kaur
- Department of Anthropology, Panjab University, Chandigarh, India
| | - M Kaur
- Department of Anthropology, Panjab University, Chandigarh, India
| |
Collapse
|
14
|
Frye HE, Izumi Y, Harris AN, Williams SB, Trousdale CR, Sun MY, Sauerbeck AD, Kummer TT, Mennerick S, Zorumski CF, Nelson EC, Dougherty JD, Morón JA. Sex Differences in the Role of CNIH3 on Spatial Memory and Synaptic Plasticity. Biol Psychiatry 2021; 90:766-780. [PMID: 34548146 PMCID: PMC8571071 DOI: 10.1016/j.biopsych.2021.07.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/12/2021] [Accepted: 07/12/2021] [Indexed: 01/23/2023]
Abstract
BACKGROUND CNIH3 is an AMPA receptor (AMPAR) auxiliary protein prominently expressed in the dorsal hippocampus (dHPC), a region that plays a critical role in spatial memory and synaptic plasticity. However, the effects of CNIH3 on AMPAR-dependent synaptic function and behavior have not been investigated. METHODS We assessed a gain-of-function model of Cnih3 overexpression in the dHPC and generated and characterized a line of Cnih3-/- C57BL/6 mice. We assessed spatial memory through behavioral assays, protein levels of AMPAR subunits and synaptic proteins by immunoblotting, and long-term potentiation in electrophysiological recordings. We also utilized a super-resolution imaging workflow, SEQUIN (Synaptic Evaluation and Quantification by Imaging of Nanostructure), for analysis of nanoscale synaptic connectivity in the dHPC. RESULTS Overexpression of Cnih3 in the dHPC improved short-term spatial memory in female mice but not in male mice. Cnih3-/- female mice exhibited weakened short-term spatial memory, reduced dHPC synapse density, enhanced expression of calcium-impermeable AMPAR (GluA2-containing) subunits in synaptosomes, and attenuated long-term potentiation maintenance compared with Cnih3+/+ control mice; Cnih3-/- males were unaffected. Further investigation revealed that deficiencies in spatial memory and changes in AMPAR composition and synaptic plasticity were most pronounced during the metestrus phase of the estrous cycle in female Cnih3-/- mice. CONCLUSIONS This study identified a novel effect of sex and estrous on CNIH3's role in spatial memory and synaptic plasticity. Manipulation of CNIH3 unmasked sexually dimorphic effects on spatial memory, synaptic function, AMPAR composition, and hippocampal plasticity. These findings reinforce the importance of considering sex as a biological variable in studies of memory and hippocampal synaptic function.
Collapse
Affiliation(s)
- Hannah E Frye
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; Pain Center, Washington University School of Medicine, St. Louis, Missouri; Program in Neuroscience, Washington University in St. Louis, St. Louis, Missouri
| | - Yukitoshi Izumi
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri; Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri
| | - Alexis N Harris
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Sidney B Williams
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; Pain Center, Washington University School of Medicine, St. Louis, Missouri
| | - Christopher R Trousdale
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; Pain Center, Washington University School of Medicine, St. Louis, Missouri
| | - Min-Yu Sun
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Andrew D Sauerbeck
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Terrance T Kummer
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Steven Mennerick
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri; Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri
| | - Charles F Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri; Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri
| | - Elliot C Nelson
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Joseph D Dougherty
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri; Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Jose A Morón
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; Pain Center, Washington University School of Medicine, St. Louis, Missouri; Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri; Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
15
|
Hormone Treatment and Alzheimer's Disease: Déjà Vu or Confused All Over Again? Drugs Aging 2021; 38:793-795. [PMID: 34342861 DOI: 10.1007/s40266-021-00887-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2021] [Indexed: 10/20/2022]
|
16
|
Prado RCR, Silveira R, Kilpatrick MW, Pires FO, Asano RY. Menstrual Cycle, Psychological Responses, and Adherence to Physical Exercise: Viewpoint of a Possible Barrier. Front Psychol 2021; 12:525943. [PMID: 33679501 PMCID: PMC7929979 DOI: 10.3389/fpsyg.2021.525943] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 01/12/2021] [Indexed: 11/29/2022] Open
Affiliation(s)
- Raul Cosme Ramos Prado
- Women's Science Studies and Research Academy, São Paulo, Brazil.,Exercise Psychophysiology Research Group, School of Arts, Sciences and Humanities, University of São Paulo, São Paulo, Brazil
| | - Rodrigo Silveira
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | | | - Flávio Oliveira Pires
- Exercise Psychophysiology Research Group, School of Arts, Sciences and Humanities, University of São Paulo, São Paulo, Brazil
| | - Ricardo Yukio Asano
- Exercise Psychophysiology Research Group, School of Arts, Sciences and Humanities, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
17
|
Kapur J, Joshi S. Progesterone modulates neuronal excitability bidirectionally. Neurosci Lett 2021; 744:135619. [PMID: 33421486 PMCID: PMC7821816 DOI: 10.1016/j.neulet.2020.135619] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/24/2020] [Accepted: 12/29/2020] [Indexed: 11/16/2022]
Abstract
Progesterone acts on neurons directly by activating its receptor and through metabolic conversion to neurosteroids. There is emerging evidence that progesterone exerts excitatory effects by activating its cognate receptors (progesterone receptors, PRs) through enhanced expression of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs). Progesterone metabolite 5α,3α-tetrahydro-progesterone (allopregnanolone, THP) mediates its anxiolytic and sedative actions through the potentiation of synaptic and extrasynaptic γ-aminobutyric acid type-A receptors (GABAARs). Here, we review progesterone's neuromodulatory actions exerted through PRs and THP and their opposing role in regulating seizures, catamenial epilepsy, and seizure exacerbation associated with progesterone withdrawal.
Collapse
Affiliation(s)
- Jaideep Kapur
- Department of Neurology, University of Virginia-HSC, Charlottesville, VA, 22908, United States; Department of Neuroscience, University of Virginia-HSC, Charlottesville, VA, 22908, United States; UVA Brain Institute, University of Virginia-HSC, Charlottesville, VA, 22908, United States
| | - Suchitra Joshi
- Department of Neurology, University of Virginia-HSC, Charlottesville, VA, 22908, United States.
| |
Collapse
|
18
|
Qu M, Wang J, Chen DC, Chen S, Xiu MH, Zhang XY. Sex-specific association between peripheral superoxide dismutase, BDNF and cognitive impairment in drug-naive first episode patients with schizophrenia. Free Radic Biol Med 2020; 160:887-893. [PMID: 32949664 DOI: 10.1016/j.freeradbiomed.2020.09.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 12/15/2022]
Abstract
Patients with schizophrenia (SCZ) have cognitive impairments across several domains. Cognition decline is related to the complex interrelationship between brain-derived neurotrophic factor (BDNF) and redox system imbalance. However, the effect of sex on cognitive impairment and biomarkers has not been fully studied in patients with drug-naïve first episode (DNFE) SCZ. 327 DNFE SCZ patients and 391 healthy controls were recruited, and the levels of BDNF and malondialdehyde (MDA) and the activities of total SOD, Mn-SOD, CuZn-SOD enzymes were measured. Cognitive function was measured by using the Repeatable Battery for the Assessment of Neuropsychological status (RBANS) and clinical symptoms by the Positive and Negative Syndrome Scale (PANSS). Patients performed worse on most cognitive tasks than controls, but there was no significant sex difference in cognitive function between patients and controls. Further analysis showed that a sex difference in MDA was found in controls rather than patients, indicating that MDA levels in men were higher than those in women in controls. Moreover, the Mn-SOD was significantly correlated with attention, language and RBANS total scores only in male patients. Multiple linear regression analysis showed that the interaction between BDNF and Mn-SOD or SOD was associated with RBANS language index score in male patients. Our results suggest that the interrelationship of BDNF with antioxidant mechanisms may contribute to the pathological mechanisms underlying cognitive deficits only in male DNFE patients with SCZ, but not in female patients.
Collapse
Affiliation(s)
- Miao Qu
- Neurology Department, Xuan Wu Hospital of Capital Medical University, Beijing, China
| | - Jun Wang
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, PR China
| | - Da Chun Chen
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, PR China
| | - Song Chen
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, PR China
| | - Mei Hong Xiu
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, PR China.
| | - Xiang Yang Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
19
|
Ghoweri AO, Ouillette L, Frazier HN, Anderson KL, Lin RL, Gant JC, Parent R, Moore S, Murphy GG, Thibault O. Electrophysiological and Imaging Calcium Biomarkers of Aging in Male and Female 5×FAD Mice. J Alzheimers Dis 2020; 78:1419-1438. [PMID: 33164928 PMCID: PMC7836067 DOI: 10.3233/jad-200109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND In animal models and tissue preparations, calcium dyshomeostasis is a biomarker of aging and Alzheimer's disease that is associated with synaptic dysfunction, neuritic pruning, and dysregulated cellular processes. It is unclear, however, whether the onset of calcium dysregulation precedes, is concurrent with, or is the product of pathological cellular events (e.g., oxidation, amyloid-β production, and neuroinflammation). Further, neuronal calcium dysregulation is not always present in animal models of amyloidogenesis, questioning its reliability as a disease biomarker. OBJECTIVE Here, we directly tested for the presence of calcium dysregulation in dorsal hippocampal neurons in male and female 5×FAD mice on a C57BL/6 genetic background using sharp electrodes coupled with Oregon-green Bapta-1 imaging. We focused on three ages that coincide with the course of amyloid deposition: 1.5, 4, and 10 months old. METHODS Outcome variables included measures of the afterhyperpolarization, short-term synaptic plasticity, and calcium kinetics during synaptic activation. Quantitative analyses of spatial learning and memory were also conducted using the Morris water maze. Main effects of sex, age, and genotype were identified on measures of electrophysiology and calcium imaging. RESULTS Measures of resting Oregon-green Bapta-1 fluorescence showed significant reductions in the 5×FAD group compared to controls. Deficits in spatial memory, along with increases in Aβ load, were detectable at older ages, allowing us to test for temporal associations with the onset of calcium dysregulation. CONCLUSION Our results provide evidence that reduced, rather than elevated, neuronal calcium is identified in this 5×FAD model and suggests that this surprising result may be a novel biomarker of AD.
Collapse
Affiliation(s)
- Adam O Ghoweri
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Lara Ouillette
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Hilaree N Frazier
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Katie L Anderson
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Ruei-Lung Lin
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - John C Gant
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Rachel Parent
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Shannon Moore
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.,5037 BSRB, Molecular and Integrative Physiology, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Geoffrey G Murphy
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.,5037 BSRB, Molecular and Integrative Physiology, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Olivier Thibault
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
20
|
Notaras M, van den Buuse M. Neurobiology of BDNF in fear memory, sensitivity to stress, and stress-related disorders. Mol Psychiatry 2020; 25:2251-2274. [PMID: 31900428 DOI: 10.1038/s41380-019-0639-2] [Citation(s) in RCA: 219] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 12/01/2019] [Accepted: 12/12/2019] [Indexed: 01/17/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is widely accepted for its involvement in resilience and antidepressant drug action, is a common genetic locus of risk for mental illnesses, and remains one of the most prominently studied molecules within psychiatry. Stress, which arguably remains the "lowest common denominator" risk factor for several mental illnesses, targets BDNF in disease-implicated brain regions and circuits. Altered stress-related responses have also been observed in animal models of BDNF deficiency in vivo, and BDNF is a common downstream intermediary for environmental factors that potentiate anxiety- and depressive-like behavior. However, BDNF's broad functionality has manifested a heterogeneous literature; likely reflecting that BDNF plays a hitherto under-recognized multifactorial role as both a regulator and target of stress hormone signaling within the brain. The role of BDNF in vulnerability to stress and stress-related disorders, such as posttraumatic stress disorder (PTSD), is a prominent example where inconsistent effects have emerged across numerous models, labs, and disciplines. In the current review we provide a contemporary update on the neurobiology of BDNF including new data from the behavioral neuroscience and neuropsychiatry literature on fear memory consolidation and extinction, stress, and PTSD. First we present an overview of recent advances in knowledge on the role of BDNF within the fear circuitry, as well as address mounting evidence whereby stress hormones interact with endogenous BDNF-TrkB signaling to alter brain homeostasis. Glucocorticoid signaling also acutely recruits BDNF to enhance the expression of fear memory. We then include observations that the functional common BDNF Val66Met polymorphism modulates stress susceptibility as well as stress-related and stress-inducible neuropsychiatric endophenotypes in both man and mouse. We conclude by proposing a BDNF stress-sensitivity hypothesis, which posits that disruption of endogenous BDNF activity by common factors (such as the BDNF Val66Met variant) potentiates sensitivity to stress and, by extension, vulnerability to stress-inducible illnesses. Thus, BDNF may induce plasticity to deleteriously promote the encoding of fear and trauma but, conversely, also enable adaptive plasticity during extinction learning to suppress PTSD-like fear responses. Ergo regulators of BDNF availability, such as the Val66Met polymorphism, may orchestrate sensitivity to stress, trauma, and risk of stress-induced disorders such as PTSD. Given an increasing interest in personalized psychiatry and clinically complex cases, this model provides a framework from which to experimentally disentangle the causal actions of BDNF in stress responses, which likely interact to potentiate, produce, and impair treatment of, stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Michael Notaras
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA.
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia. .,College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia. .,Department of Pharmacology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
21
|
Acharya KD, Nettles SA, Lichti CF, Warre-Cornish K, Polit LD, Srivastava DP, Denner L, Tetel MJ. Dopamine-induced interactions of female mouse hypothalamic proteins with progestin receptor-A in the absence of hormone. J Neuroendocrinol 2020; 32:e12904. [PMID: 33000549 PMCID: PMC7591852 DOI: 10.1111/jne.12904] [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: 04/22/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 11/26/2022]
Abstract
Neural progestin receptors (PR) function in reproduction, neural development, neuroprotection, learning, memory and the anxiety response. In the absence of progestins, PR can be activated by dopamine (DA) in the rodent hypothalamus to elicit female sexual behaviour. The present study investigated mechanisms of DA activation of PR by testing the hypothesis that proteins from DA-treated hypothalami interact with PR in the absence of progestins. Ovariectomised, oestradiol-primed mice were infused with a D1-receptor agonist, SKF38393 (SKF), into the third ventricle 30 minutes prior to death. Proteins from SKF-treated hypothalami were pulled-down with glutathione S-transferase-tagged mouse PR-A or PR-B and the interactomes were analysed by mass spectrometry. The largest functional group to interact with PR-A in a DA-dependent manner was synaptic proteins. To test the hypothesis that DA activation of PR regulates synaptic proteins, we developed oestradiol-induced PR-expressing hypothalamic-like neurones derived from human-induced pluripotent stem cells (hiPSCs). Similar to progesterone (P4), SKF treatment of hiPSCs increased synapsin1/2 expression. This SKF-dependent effect was blocked by the PR antagonist RU486, suggesting that PR are necessary for this DA-induced increase. The second largest DA-dependent PR-A protein interactome comprised metabolic regulators involved in glucose metabolism, lipid synthesis and mitochondrial energy production. Interestingly, hypothalamic proteins interacted with PR-A, but not PR-B, in an SKF-dependent manner, suggesting that DA promotes the interaction of multiple hypothalamic proteins with PR-A. These in vivo and in vitro results indicate novel mechanisms by which DA can differentially activate PR isoforms in the absence of P4 and provide a better understanding of ligand-independent PR activation in reproductive, metabolic and mental health disorders in women.
Collapse
Affiliation(s)
| | | | - Cheryl F. Lichti
- Department of Pathology and Immunology, Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO 63110
| | - Katherine Warre-Cornish
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, SE5 8AF, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| | - Lucia Dutan Polit
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, SE5 8AF, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, SE5 8AF, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| | - Larry Denner
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Marc J. Tetel
- Neuroscience Department, Wellesley College, Wellesley, MA 02481
| |
Collapse
|
22
|
Proaño SB, Krentzel AA, Meitzen J. Differential and synergistic roles of 17β-estradiol and progesterone in modulating adult female rat nucleus accumbens core medium spiny neuron electrophysiology. J Neurophysiol 2020; 123:2390-2405. [PMID: 32401164 DOI: 10.1152/jn.00157.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Naturally occurring cyclical changes in sex steroid hormones such as 17β-estradiol and progesterone can modulate neuron function and behavior in female mammals. One example is the estrous cycle in rats, which is composed of multiple phases. We previously reported evidence of differences between estrous cycle phases in excitatory synapse and intrinsic electrophysiological properties of rat nucleus accumbens core (AcbC) medium spiny neurons (MSNs). The AcbC is a nexus between the limbic and premotor systems and is integral for controlling motivated and reward-associated behaviors and disorders, which are sensitive to the estrous cycle and hormones. The present study expands our prior findings by testing whether circulating levels of estradiol and progesterone correlate with changes in MSN electrophysiology across estrous cycle phases. As part of this project, the excitatory synapse and intrinsic excitability properties of MSNs in late proestrus of adult female rats were assessed. Circulating levels of estradiol correlate with resting membrane potential, the time constant of the membrane, and rheobase. Circulating levels of progesterone correlate with miniature excitatory postsynaptic current (mEPSC) frequency and amplitude. Circulating levels of estradiol and progesterone together correlate with mEPSC amplitude, resting membrane potential, and input resistance. The late proestrus phase features a prominent and unique decrease in mEPSC frequency. These data indicate that circulating levels of estradiol and progesterone alone or in combination interact with specific MSN electrophysiological properties, indicating differential and synergistic roles of these hormones. Broadly, these findings illustrate the underlying endocrine actions regarding how the estrous cycle modulates MSN electrophysiology.NEW & NOTEWORTHY This research indicates that estradiol and progesterone act both differentially and synergistically to modulate neuron physiology in the nucleus accumbens core. These actions by specific hormones provide key data indicating the endocrine mechanisms underlying how the estrous cycle modulates neuron physiology in this region. Overall, these data reinforce that hormones are an important influence on neural physiology.
Collapse
Affiliation(s)
- Stephanie B Proaño
- Graduate Program in Biology, North Carolina State University, Raleigh, North Carolina.,W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina.,Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina
| | - Amanda A Krentzel
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina.,Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina
| | - John Meitzen
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina.,Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina.,Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
23
|
Cáceres ARR, Vega Orozco AS, Cabrera RJ, Laconi MR. "Rapid actions of the neurosteroid allopregnanolone on ovarian and hypothalamic steroidogenesis: Central and peripheral modulation". J Neuroendocrinol 2020; 32:e12836. [PMID: 32062869 DOI: 10.1111/jne.12836] [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: 07/17/2019] [Revised: 01/24/2020] [Accepted: 01/24/2020] [Indexed: 11/28/2022]
Abstract
The present study aimed to determine whether an i.c.v. administration of allopregnanolone (ALLO) rapidly modifies the hypothalamic and ovarian 3β-hydroxysteroid dehydrogenase (3β-HSD) enzymatic activity and gene expression in in vivo and ex vivo systems in pro-oestrus (PE) and dioestrus I (DI) rats. Animals were injected with vehicle, ALLO, bicuculline or bicuculline plus ALLO and were then killed. In the in vivo experiment, the hypothalamus, ovaries and serum were extracted and analysed. In the ex vivo experiment, the superior mesenteric ganglion - ovarian nerve plexus - ovary system was extracted and incubated during 120 minutes at 37 ºC. The serum and ovarian compartment fluids were used to determine progesterone by radioimmunoanalysis. In the in vivo experiments, ALLO caused a decrease in hypothalamic and ovarian 3β-HSD enzymatic activity during PE. During DI, ALLO increased hypothalamic and ovarian 3β-HSD activity and gene expression. The ovarian 3β-HSD activity increased in both stages in the ex vivo system; gene expression increased only during DI. ALLO induced an increase in serum progesterone only in D1 and in the ovarian incubation liquids in both stages. All findings were reversed by an injection of bicuculline before ALLO. Ovarian steroidogenic changes could be attributed to signals coming from ganglion neurones, which are affected by the acute central neurosteroid stimulation. The i.c.v. administration of ALLO via the GABAergic system altered 3β-HSD activity and gene expression, modulating the neuroendocrine axis. The present study reveals the action that ALLO exerts on the GABAA receptor in both the central and peripheral nervous system and its relationship with hormonal variations. ALLO is involved in the "fine tuning" of neurosecretory functions as a potent modulator of reproductive processes in female rats.
Collapse
Affiliation(s)
- Antonella Rosario Ramona Cáceres
- Laboratorio de Fisiopatología Ovárica, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU - CONICET Mendoza), Mendoza, Argentina
- Facultad de Ingeniería y Facultad de Ciencias Médicas, Universidad de Mendoza, Mendoza, Argentina
- Facultad de Ciencias Veterinarias y Ambientales, Universidad Juan Agustín Maza, Mendoza, Argentina
| | - Adriana Soledad Vega Orozco
- Laboratorio de Biología de la Reproducción (LABIR), Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
- Facultad de Ciencias de la Salud, Instituto de Investigaciones Biomédicas, Universidad de Mendoza (INBIOMED-IMBECU - CONICET), Mendoza, Argentina
| | - Ricardo Jorge Cabrera
- Facultad de Ciencias de la Salud, Instituto de Investigaciones Biomédicas, Universidad de Mendoza (INBIOMED-IMBECU - CONICET), Mendoza, Argentina
| | - Myriam Raquel Laconi
- Laboratorio de Fisiopatología Ovárica, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU - CONICET Mendoza), Mendoza, Argentina
- Facultad de Ingeniería y Facultad de Ciencias Médicas, Universidad de Mendoza, Mendoza, Argentina
| |
Collapse
|
24
|
Giatti S, Diviccaro S, Serafini MM, Caruso D, Garcia-Segura LM, Viviani B, Melcangi RC. Sex differences in steroid levels and steroidogenesis in the nervous system: Physiopathological role. Front Neuroendocrinol 2020; 56:100804. [PMID: 31689419 DOI: 10.1016/j.yfrne.2019.100804] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/10/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022]
Abstract
The nervous system, in addition to be a target for steroid hormones, is the source of a variety of neuroactive steroids, which are synthesized and metabolized by neurons and glial cells. Recent evidence indicates that the expression of neurosteroidogenic proteins and enzymes and the levels of neuroactive steroids are different in the nervous system of males and females. We here summarized the state of the art of neuroactive steroids, particularly taking in consideration sex differences occurring in the synthesis and levels of these molecules. In addition, we discuss the consequences of sex differences in neurosteroidogenesis for the function of the nervous system under healthy and pathological conditions and the implications of neuroactive steroids and neurosteroidogenesis for the development of sex-specific therapeutic interventions.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Melania Maria Serafini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Donatella Caruso
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, 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
| | - Barbara Viviani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Roberto C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
25
|
Ratnakumar A, Zimmerman SE, Jordan BA, Mar JC. Estrogen activates Alzheimer's disease genes. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:906-917. [PMID: 31890855 PMCID: PMC6926344 DOI: 10.1016/j.trci.2019.09.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Introduction Women are at increased risk for Alzheimer's disease (AD), but the reason why remains unknown. One hypothesis is that low estrogen levels at menopause increases vulnerability to AD, but this remains unproven. Methods We compared neuronal genes upregulated by estrogen in ovariectomized female rhesus macaques with a database of >17,000 diverse gene sets and applied a rare variant burden test to exome sequencing data from 1208 female AD patients with the age of onset < 75 years and 2162 female AD controls. Results We found a striking overlap between genes upregulated by estrogen in macaques and genes downregulated in the human postmortem AD brain, and we found that estrogen upregulates the APOE gene and that progesterone acts antagonistically to estrogen genome-wide. We also found that female patients with AD have excess rare mutations in the early menopause gene MCM8. Discussion We show with genomic data that the menopausal loss of estrogen could underlie the increased risk for AD in women.
Collapse
Affiliation(s)
- Abhirami Ratnakumar
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Samuel E Zimmerman
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bryen A Jordan
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jessica C Mar
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA.,Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Australia
| |
Collapse
|
26
|
Kelicen-Ugur P, Cincioğlu-Palabıyık M, Çelik H, Karahan H. Interactions of Aromatase and Seladin-1: A Neurosteroidogenic and Gender Perspective. Transl Neurosci 2019; 10:264-279. [PMID: 31737354 PMCID: PMC6843488 DOI: 10.1515/tnsci-2019-0043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 10/03/2019] [Indexed: 12/16/2022] Open
Abstract
Aromatase and seladin-1 are enzymes that have major roles in estrogen synthesis and are important in both brain physiology and pathology. Aromatase is the key enzyme that catalyzes estrogen biosynthesis from androgen precursors and regulates the brain’s neurosteroidogenic activity. Seladin-1 is the enzyme that catalyzes the last step in the biosynthesis of cholesterol, the precursor of all hormones, from desmosterol. Studies indicated that seladin-1 is a downstream mediator of the neuroprotective activity of estrogen. Recently, we also showed that there is an interaction between aromatase and seladin-1 in the brain. Therefore, the expression of local brain aromatase and seladin-1 is important, as they produce neuroactive steroids in the brain for the protection of neuronal damage. Increasing steroid biosynthesis specifically in the central nervous system (CNS) without affecting peripheral hormone levels may be possible by manipulating brain-specific promoters of steroidogenic enzymes. This review emphasizes that local estrogen, rather than plasma estrogen, may be responsible for estrogens’ protective effects in the brain. Therefore, the roles of aromatase and seladin-1 and their interactions in neurodegenerative events such as Alzheimer’s disease (AD), ischemia/reperfusion injury (stroke), and epilepsy are also discussed in this review.
Collapse
Affiliation(s)
- Pelin Kelicen-Ugur
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, Sıhhiye Ankara Turkey
| | - Mehtap Cincioğlu-Palabıyık
- Turkish Medicines and Medical Devices Agency (TITCK), Department of Regulatory Affairs, Division of Pharmacological Assessment, Ankara, Turkey
| | - Hande Çelik
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, Sıhhiye Ankara Turkey
| | - Hande Karahan
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
27
|
Roney JR. On the use of log transformations when testing hormonal predictors of cycle phase shifts: Commentary on. EVOL HUM BEHAV 2019. [DOI: 10.1016/j.evolhumbehav.2019.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
28
|
Hillerer KM, Slattery DA, Pletzer B. Neurobiological mechanisms underlying sex-related differences in stress-related disorders: Effects of neuroactive steroids on the hippocampus. Front Neuroendocrinol 2019; 55:100796. [PMID: 31580837 PMCID: PMC7115954 DOI: 10.1016/j.yfrne.2019.100796] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022]
Abstract
Men and women differ in their vulnerability to a variety of stress-related illnesses, but the underlying neurobiological mechanisms are not well understood. This is likely due to a comparative dearth of neurobiological studies that assess male and female rodents at the same time, while human neuroimaging studies often don't model sex as a variable of interest. These sex differences are often attributed to the actions of sex hormones, i.e. estrogens, progestogens and androgens. In this review, we summarize the results on sex hormone actions in the hippocampus and seek to bridge the gap between animal models and findings in humans. However, while effects of sex hormones on the hippocampus are largely consistent in animals and humans, methodological differences challenge the comparability of animal and human studies on stress effects. We summarise our current understanding of the neurobiological mechanisms that underlie sex-related differences in behavior and discuss implications for stress-related illnesses.
Collapse
Affiliation(s)
- Katharina M Hillerer
- Department of Obstetrics and Gynaecology, Salzburger Landeskrankenhaus (SALK), Paracelsus Medical University (PMU), Clinical Research Center Salzburg (CRCS), Salzburg, Austria.
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Belinda Pletzer
- Department of Psychology, University of Salzburg, Salzburg, Austria; Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria
| |
Collapse
|
29
|
Tronson NC, Keiser AA. A Dynamic Memory Systems Framework for Sex Differences in Fear Memory. Trends Neurosci 2019; 42:680-692. [PMID: 31473031 DOI: 10.1016/j.tins.2019.07.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 06/17/2019] [Accepted: 07/29/2019] [Indexed: 01/04/2023]
Abstract
Emerging research demonstrates that a pattern of overlapping but distinct molecular and circuit mechanisms are engaged by males and females during memory tasks. Importantly, sex differences in neural mechanisms and behavioral strategies are evident even when performance on a memory task is similar between females and males. We propose that sex differences in memory may be best understood within a dynamic memory systems framework. Specifically, sex differences in hormonal influences and neural circuit development result in biases in the circuits engaged and the information preferentially stored or retrieved in males and females. By using animal models to understand the neural networks and molecular mechanisms required for memory in both sexes, we can gain crucial insights into sex and gender biases in disorders including post-traumatic stress disorder (PTSD) in humans.
Collapse
Affiliation(s)
- Natalie C Tronson
- Psychology Department, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Ashley A Keiser
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| |
Collapse
|
30
|
Age and sex differences in the innate defensive behaviors of C57BL/6 mice exhibited in a fear conditioning paradigm and upon exposure to a predatory odor. Physiol Behav 2019; 204:264-274. [PMID: 30840847 DOI: 10.1016/j.physbeh.2019.02.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/23/2019] [Accepted: 02/23/2019] [Indexed: 12/29/2022]
Abstract
Defensive responses of mice include an array of specific behaviors that involve adaptive components based on the assessment of the threat of confrontation. The freezing response is represented by a motionless moment without any specific posture or behavioral sequence, and it is widely used in the fear conditioning paradigms and other relevant defensive situations. However, freezing measurements include fragmental components of several defensive behaviors that are exhibited during the session, such as behavioral inhibition, crouching, and a moment of risk assessment behavior. From an ethological view, behavioral analyses of C57BL/6 mice of both sexes and three different ages (postnatal days (P) 25, 35, and 65) revealed a rich variety of defensive behaviors during a fear conditioning session and in response to predatory odor exposure as a nonconditioned behavior. P-25 and 35 mice exhibited more behavioral inhibition than P-65 adult mice, and P-65 mice exhibited a crouching posture more often than younger mice. This age difference was more pronounced in males. The stretch-attend posture (SAP) increased with age, except in P-25 males, which exhibited robust SAP in response to a conditioned cue; this response indicates that P-25 males are defensive in a situation-nonmatching manner. Situation-dependent defense strategies were revealed in P-35 and 65 mice: Fear conditioning paradigm induced more robust defensive responses than predatory odor exposure, to which mice primarily exhibited SAP. A sex-based difference was revealed in adult mice. Males tended to show more passive defensive responses, such as crouching and withdrawal, and females exhibited more active responses, such as SAP. These age- and sex-based differences may stem from the ethological demands and illustrate adolescent ontogenetic processes of defense behavior.
Collapse
|
31
|
Zhang YY, Liu MY, Liu Z, Zhao JK, Zhao YG, He L, Li W, Zhang JQ. GPR30-mediated estrogenic regulation of actin polymerization and spatial memory involves SRC-1 and PI3K-mTORC2 in the hippocampus of female mice. CNS Neurosci Ther 2019; 25:714-733. [PMID: 30714337 PMCID: PMC6515707 DOI: 10.1111/cns.13108] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/18/2018] [Accepted: 01/03/2019] [Indexed: 12/29/2022] Open
Abstract
AIMS The G-protein-coupled estrogen receptor GPR30 (also referred to as GPER) has been implicated in the estrogenic regulation of hippocampal plasticity and spatial memory; however, the molecular mechanisms are largely unclear. METHODS In this study, we initially examined the levels of GPR30 in the hippocampus of postnatal, ovariectomy (OVX)- and letrozole (LET)-treated female mice. Under G1, G15, and/or OVX treatment, the spatial memory, spine density, levels of ERα, ERβ, and SRC-1, selected synaptic proteins, mTORC2 signals (Rictor and p-AKT Ser473), and actin polymerization dynamics were subsequently evaluated. Furthermore, G1, G15, and/or E2 combined with SRC-1 and/or PI3K inhibitors, actin cytoskeleton polymerization modulator JPK, and CytoD treatments were used to address the mechanisms that underlie GPR30 regulation in vitro. Finally, mTORC2 activator A-443654 (A4) was used to explore the role of mTORC2 in GPR30 regulation of spatial memory. RESULTS The results showed that high levels of GPR30 were detected in the adult hippocampus and the levels were downregulated by OVX and LET. OVX induced an impairment of spatial memory, and changes in other parameters previously described were reversed by G1 and mimicked by G15. Furthermore, the E2 effects on SRC-1 and mTORC2 signals, synaptic proteins, and actin polymerization were inhibited by G15, whereas G1 effects on these parameters were inhibited by the blockade of SRC-1 or PI3K; the levels of synaptic proteins were regulated by JPK and CytoD. Importantly, G15-induced actin depolymerization and spatial memory impairment were rescued by mTORC2 activation with A4. CONCLUSIONS Taking together, these results demonstrated that decreased GPR30 induces actin depolymerization through SRC-1 and PI3K/mTORC2 pathways and ultimately impairs learning and memory, indicating its potential role as a therapeutic target against hippocampus-based, E2-related memory impairments.
Collapse
Affiliation(s)
- Yuan-Yuan Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing, China
| | - Meng-Ying Liu
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing, China
| | - Zhi Liu
- Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Ji-Kai Zhao
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing, China
| | - Yan-Gang Zhao
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing, China
| | - Li He
- School of Nursing, Third Military Medical University, Chongqing, China
| | - Wei Li
- School of Nursing, Third Military Medical University, Chongqing, China
| | - Ji-Qiang Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing, China
| |
Collapse
|
32
|
Moore AM, Abbott G, Mair J, Prescott M, Campbell RE. Mapping GABA and glutamate inputs to gonadotrophin-releasing hormone neurones in male and female mice. J Neuroendocrinol 2018; 30:e12657. [PMID: 30415474 DOI: 10.1111/jne.12657] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/22/2018] [Accepted: 11/05/2018] [Indexed: 11/28/2022]
Abstract
Gonadotrophin-releasing hormone (GnRH) neurone function is dependent upon gonadal steroid hormone feedback, which is communicated in large part through an afferent neuronal network. The classical neurotransmitters GABA and glutamate are important regulators of GnRH neurone activity and are implicated in mediating feedback signals. In the present study, we aimed to determine whether GABAergic or glutamatergic input to GnRH neurones differs between males and females and/or exhibits morphological plasticity in response to steroid hormone feedback in females. Tissue collected from GnRH-green fluorescent protein (GFP) male and female mice in dioestrus underwent immunofluorescence labelling of GFP and either the vesicular GABA transporter (VGAT) or the vesicular glutamate transporter 2 (VGLUT2). No differences in the densities or absolute numbers of VGAT-immunoreactive (-IR) or VGLUT2-IR puncta apposed to GnRH neurones were identified between males and females. The most significant input from either neurotransmitter was to the proximal dendritic region and 80% of VGAT-IR puncta apposed to GnRH neurones co-localised with synaptophysin. Putative inputs were also assessed in ovariectomised (OVX) female mice treated with negative (OVX+E) or positive (OVX+E+E) feedback levels of oestrogen, and OVX+E+E mice were killed during the expected GnRH/luteinising hormone surge. No differences in VGLUT2-IR contacts to GnRH neurones were identified between animals under the negative-feedback influence of oestrogen (OVX+E) or the positive influence of oestrogen (OVX+E+E), regardless of cFos activation status. By contrast, a significant elevation in putative GABAergic inputs to GnRH neurones at the time of the preovulatory surge was found in the cFos-negative subset of GnRH neurones, both at the level of the soma and at the proximal dendrite. Taken together, these data suggest that, although GABAergic and glutamatergic innervation of GnRH neurones is not sexually differentiated, cyclic fluctuations in steroid hormone feedback over the female oestrous cycle result in plastic changes in GABAergic inputs to a subpopulation of GnRH neurones.
Collapse
Affiliation(s)
- Aleisha M Moore
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
| | - Georgina Abbott
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Jonathan Mair
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Melanie Prescott
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
33
|
Progesterone, 5a-dihydropogesterone and allopregnanolone's effects on seizures: A review of animal and clinical studies. Seizure 2018; 63:26-36. [DOI: 10.1016/j.seizure.2018.10.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/16/2018] [Accepted: 10/20/2018] [Indexed: 02/07/2023] Open
|
34
|
Hoffman MA, Doeringer JR, Norcross MF, Johnson ST, Chappell PE. Presynaptic inhibition decreases when estrogen level rises. Scand J Med Sci Sports 2018; 28:2009-2015. [PMID: 29744948 DOI: 10.1111/sms.13210] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2018] [Indexed: 12/14/2022]
Abstract
The objective was to determine estrogen's influence on control of a skeletal muscle through measurements of motorneuron excitability (H:M ratio) and presynaptic inhibition (PI). Estrogen serum concentrations were measured at menses and ovulation of female subjects and compared to male controls. Data were analyzed from 12 women and 13 men reporting no history of knee ligament injury. Women reported regular menstrual cycles and no hormone-based contraceptive use for the previous year. Women were tested at menses (Time1) and ovulation (Time2). Men were tested twice, approximately 14 days apart. Analysis indicated no difference in the H:M ratio between the sexes at either time point. A significant difference for the sexes was detected in the magnitude of estrogen change (∆EST) between observations. At Time1, the male and female estrogen concentrations were not different; however, they were different at Time2, primarily due to the large rise observed in the women. A significant difference between the sexes was also seen in the magnitude of change for PI (∆PI) between observations. As with EST, the levels of PI between the sexes at Time1 were not different; however, a difference existed at Time 2. Estrogen interacts with GABA at several nervous system locations affecting inhibition of synaptic transmission. This is the first study to investigate changes in PI of a skeletal muscle between times of low and high estrogen. Improving the understanding of estrogen's influence on skeletal muscles may provide answers to why noncontact anterior cruciate ligament injuries of the knee occur more frequently in women.
Collapse
Affiliation(s)
| | - J R Doeringer
- Nova Southeastern University, Fort Lauderdale, FL, USA
| | | | | | | |
Collapse
|
35
|
Joshi S, Sun H, Rajasekaran K, Williamson J, Perez-Reyes E, Kapur J. A novel therapeutic approach for treatment of catamenial epilepsy. Neurobiol Dis 2018; 111:127-137. [PMID: 29274741 PMCID: PMC5803337 DOI: 10.1016/j.nbd.2017.12.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/27/2017] [Accepted: 12/19/2017] [Indexed: 12/21/2022] Open
Abstract
Many women with epilepsy experience perimenstrual seizure exacerbation, referred to as catamenial epilepsy. There is no effective treatment for this condition, proposed to result from withdrawal of neurosteroid-mediated effects of progesterone. A double-blind, multicenter, phase III, clinical trial of catamenial epilepsy has failed to find a beneficial effect of progesterone. The neurosteroid-mediated effects of progesterone have been extensively studied in relation to catamenial epilepsy; however, the effects mediated by progesterone receptor activation have been overlooked. We determined whether progesterone increased excitatory transmission in the hippocampus via activation of progesterone receptors, which may play a role in regulating catamenial seizure exacerbation. In a double-blind study using a rat model of catamenial epilepsy, we found that treatment with RU-486, which blocks progesterone and glucocorticoid receptors, significantly attenuated neurosteroid withdrawal-induced seizures. Furthermore, progesterone treatment as well as endogenous rise in progesterone during estrous cycle increased the expression of GluA1 and GluA2 subunits of AMPA receptors in the hippocampi, and enhanced the AMPA receptor-mediated synaptic transmission of CA1 pyramidal neurons. The progesterone-induced plasticity of AMPA receptors was blocked by RU-486 treatment and progesterone also failed to increase AMPA receptor expression in progesterone receptor knockout mice. These studies demonstrate that progesterone receptor activation regulates AMPA receptor expression and may play a role in catamenial seizure exacerbation.
Collapse
Affiliation(s)
- Suchitra Joshi
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, United States
| | - Huayu Sun
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, United States
| | - Karthik Rajasekaran
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, United States
| | - John Williamson
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, United States
| | - Edward Perez-Reyes
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, United States
| | - Jaideep Kapur
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, United States; Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, United States.
| |
Collapse
|
36
|
Diotel N, Charlier TD, Lefebvre d'Hellencourt C, Couret D, Trudeau VL, Nicolau JC, Meilhac O, Kah O, Pellegrini E. Steroid Transport, Local Synthesis, and Signaling within the Brain: Roles in Neurogenesis, Neuroprotection, and Sexual Behaviors. Front Neurosci 2018; 12:84. [PMID: 29515356 PMCID: PMC5826223 DOI: 10.3389/fnins.2018.00084] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/02/2018] [Indexed: 01/18/2023] Open
Abstract
Sex steroid hormones are synthesized from cholesterol and exert pleiotropic effects notably in the central nervous system. Pioneering studies from Baulieu and colleagues have suggested that steroids are also locally-synthesized in the brain. Such steroids, called neurosteroids, can rapidly modulate neuronal excitability and functions, brain plasticity, and behavior. Accumulating data obtained on a wide variety of species demonstrate that neurosteroidogenesis is an evolutionary conserved feature across fish, birds, and mammals. In this review, we will first document neurosteroidogenesis and steroid signaling for estrogens, progestagens, and androgens in the brain of teleost fish, birds, and mammals. We will next consider the effects of sex steroids in homeostatic and regenerative neurogenesis, in neuroprotection, and in sexual behaviors. In a last part, we will discuss the transport of steroids and lipoproteins from the periphery within the brain (and vice-versa) and document their effects on the blood-brain barrier (BBB) permeability and on neuroprotection. We will emphasize the potential interaction between lipoproteins and sex steroids, addressing the beneficial effects of steroids and lipoproteins, particularly HDL-cholesterol, against the breakdown of the BBB reported to occur during brain ischemic stroke. We will consequently highlight the potential anti-inflammatory, anti-oxidant, and neuroprotective properties of sex steroid and lipoproteins, these latest improving cholesterol and steroid ester transport within the brain after insults.
Collapse
Affiliation(s)
- Nicolas Diotel
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
| | - Thierry D. Charlier
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Christian Lefebvre d'Hellencourt
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
| | - David Couret
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
- CHU de La Réunion, Saint-Denis, France
| | | | - Joel C. Nicolau
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Olivier Meilhac
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
- CHU de La Réunion, Saint-Denis, France
| | - Olivier Kah
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Elisabeth Pellegrini
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| |
Collapse
|
37
|
Moraga‐Amaro R, van Waarde A, Doorduin J, de Vries EFJ. Sex steroid hormones and brain function: PET imaging as a tool for research. J Neuroendocrinol 2018; 30:e12565. [PMID: 29237239 PMCID: PMC5838537 DOI: 10.1111/jne.12565] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/26/2017] [Accepted: 12/06/2017] [Indexed: 12/15/2022]
Abstract
Sex steroid hormones are major regulators of sexual characteristic among species. These hormones, however, are also produced in the brain. Steroidal hormone-mediated signalling via the corresponding hormone receptors can influence brain function at the cellular level and thus affect behaviour and higher brain functions. Altered steroid hormone signalling has been associated with psychiatric disorders, such as anxiety and depression. Neurosteroids are also considered to have a neuroprotective effect in neurodegenerative diseases. So far, the role of steroid hormone receptors in physiological and pathological conditions has mainly been investigated post mortem on animal or human brain tissues. To study the dynamic interplay between sex steroids, their receptors, brain function and behaviour in psychiatric and neurological disorders in a longitudinal manner, however, non-invasive techniques are needed. Positron emission tomography (PET) is a non-invasive imaging tool that is used to quantitatively investigate a variety of physiological and biochemical parameters in vivo. PET uses radiotracers aimed at a specific target (eg, receptor, enzyme, transporter) to visualise the processes of interest. In this review, we discuss the current status of the use of PET imaging for studying sex steroid hormones in the brain. So far, PET has mainly been investigated as a tool to measure (changes in) sex hormone receptor expression in the brain, to measure a key enzyme in the steroid synthesis pathway (aromatase) and to evaluate the effects of hormonal treatment by imaging specific downstream processes in the brain. Although validated radiotracers for a number of targets are still warranted, PET can already be a useful technique for steroid hormone research and facilitate the translation of interesting findings in animal studies to clinical trials in patients.
Collapse
Affiliation(s)
- R. Moraga‐Amaro
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - A. van Waarde
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - J. Doorduin
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - E. F. J. de Vries
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| |
Collapse
|
38
|
Meyer CE, Kurth F, Lepore S, Gao JL, Johnsonbaugh H, Oberoi MR, Sawiak SJ, MacKenzie-Graham A. In vivo magnetic resonance images reveal neuroanatomical sex differences through the application of voxel-based morphometry in C57BL/6 mice. Neuroimage 2017; 163:197-205. [PMID: 28923275 PMCID: PMC5716897 DOI: 10.1016/j.neuroimage.2017.09.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 09/07/2017] [Accepted: 09/14/2017] [Indexed: 12/22/2022] Open
Abstract
Behaviorally relevant sex differences are often associated with structural differences in the brain and many diseases are sexually dimorphic in prevalence and progression. Characterizing sex differences is imperative to gaining a complete understanding of behavior and disease which will, in turn, allow for a balanced approach to scientific research and the development of therapies. In this study, we generated novel tissue probability maps (TPMs) based on 30 male and 30 female in vivo C57BL/6 mouse brain magnetic resonance images and used voxel-based morphometry (VBM) to analyze sex differences. Females displayed larger anterior hippocampus, basolateral amygdala, and lateral cerebellar cortex volumes, while males exhibited larger cerebral cortex, medial amygdala, and medial cerebellar cortex volumes. Atlas-based morphometry (ABM) revealed a statistically significant sex difference in cortical volume and no difference in whole cerebellar volume. This validated our VBM findings that showed a larger cerebral cortex in male mice and a pattern of dimorphism in the cerebellum where the lateral portion was larger in females and the medial portion was larger in males. These results are consonant with previous ex vivo studies examining sex differences, but also suggest further regions of interest.
Collapse
Affiliation(s)
- Cassandra E Meyer
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, 635 Charles Young Drive South, Los Angeles, CA, USA
| | - Florian Kurth
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, 635 Charles Young Drive South, Los Angeles, CA, USA
| | - Stefano Lepore
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, 635 Charles Young Drive South, Los Angeles, CA, USA
| | - Josephine L Gao
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, 635 Charles Young Drive South, Los Angeles, CA, USA
| | - Hadley Johnsonbaugh
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, 635 Charles Young Drive South, Los Angeles, CA, USA
| | - Mandavi R Oberoi
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, 635 Charles Young Drive South, Los Angeles, CA, USA
| | - Stephen J Sawiak
- Wolfson Brain Imaging Centre, University of Cambridge, Box 65 Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Allan MacKenzie-Graham
- Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, 635 Charles Young Drive South, Los Angeles, CA, USA.
| |
Collapse
|
39
|
The Progestin Receptor Interactome in the Female Mouse Hypothalamus: Interactions with Synaptic Proteins Are Isoform Specific and Ligand Dependent. eNeuro 2017; 4:eN-NWR-0272-17. [PMID: 28955722 PMCID: PMC5605756 DOI: 10.1523/eneuro.0272-17.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 08/31/2017] [Accepted: 09/01/2017] [Indexed: 01/06/2023] Open
Abstract
Progestins bind to the progestin receptor (PR) isoforms, PR-A and PR-B, in brain to influence development, female reproduction, anxiety, and stress. Hormone-activated PRs associate with multiple proteins to form functional complexes. In the present study, proteins from female mouse hypothalamus that associate with PR were isolated using affinity pull-down assays with glutathione S-transferase–tagged mouse PR-A and PR-B. Using complementary proteomics approaches, reverse phase protein array (RPPA) and mass spectrometry, we identified hypothalamic proteins that interact with PR in a ligand-dependent and isoform-specific manner and were confirmed by Western blot. Synaptic proteins, including synapsin-I and synapsin-II, interacted with agonist-bound PR isoforms, suggesting that both isoforms function in synaptic plasticity. In further support, synaptogyrin-III and synapsin-III associated with PR-A and PR-B, respectively. PR also interacted with kinases, including c-Src, mTOR, and MAPK1, confirming phosphorylation as an integral process in rapid effects of PR in the brain. Consistent with a role in transcriptional regulation, PR associated with transcription factors and coactivators in a ligand-specific and isoform-dependent manner. Interestingly, both PR isoforms associated with a key regulator of energy homeostasis, FoxO1, suggesting a novel role for PR in energy metabolism. Because many identified proteins in this PR interactome are synaptic proteins, we tested the hypothesis that progestins function in synaptic plasticity. Indeed, progesterone enhanced synaptic density, by increasing synapsin-I–positive synapses, in rat primary cortical neuronal cultures. This novel combination of RPPA and mass spectrometry allowed identification of PR action in synaptic remodeling and energy homeostasis and reveals unique roles for progestins in brain function and disease.
Collapse
|
40
|
Nongenomic Actions of 17-β Estradiol Restore Respiratory Neuroplasticity in Young Ovariectomized Female Rats. J Neurosci 2017; 37:6648-6660. [PMID: 28592693 DOI: 10.1523/jneurosci.0433-17.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/18/2017] [Accepted: 05/05/2017] [Indexed: 12/31/2022] Open
Abstract
Gonadal steroids modulate CNS plasticity, including phrenic long-term facilitation (pLTF), a form of spinal respiratory neuroplasticity resulting in increased phrenic nerve motor output following exposure to acute intermittent hypoxia (aIH; three 5 min episodes, 10.5% O2). Despite the importance of respiratory system neuroplasticity, and its dependence on estrogen in males, little is known about pLTF expression or mechanisms of estrogen signaling in females. Here, we tested the hypotheses that (1) pLTF expression in young, gonadally intact female rats would be expressed during estrous cycle stages in which 17β-estradiol (E2) is naturally high (e.g., proestrus vs estrus), (2) pLTF would be absent in ovariectomized (OVX) rats and in physiological conditions in which serum progesterone, but not E2, is elevated (e.g., lactating rats, 3-10 d postpartum), and (3) acute E2 administration would be sufficient to restore pLTF in OVX rats. Recordings of phrenic nerve activity in female Sprague Dawley rats (3-4 months) revealed a direct correlation between serum E2 levels and pLTF expression in cycling female rats. pLTF was abolished with OVX, but was re-established by acute E2 replacement (3 h, intraperitoneal). To identify underlying E2 signaling mechanisms, we intrathecally applied BSA-conjugated E2 over the spinal phrenic motor nucleus and found that pLTF expression was restored within 15 min, suggesting nongenomic E2 effects at membrane estrogen receptors. These data are the first to investigate the role of ovarian E2 in young cycling females, and to identify a role for nongenomic estrogen signaling in any form of respiratory system neuroplasticity.SIGNIFICANCE STATEMENT Exposure to acute intermittent hypoxia induces phrenic long-term facilitation (pLTF), a form of spinal respiratory motor plasticity that improves breathing in models of spinal cord injury. Although pathways leading to pLTF are well studied in males and estradiol (E2) is known to be required, it has seldom been investigated in females, and underlying mechanisms of E2 signaling are unknown in either sex. We found that while ovariectomy abolished pLTF, it could be restored by acute systemic E2, or by intraspinal application of the membrane-impermeable E2 (BSA-conjugated E2; 15 min). The ability of nongenomic estrogen signaling within the cervical spinal cord to recover respiratory neuroplasticity in disorders of respiratory insufficiency suggests that membrane estrogen receptors may represent novel therapeutic targets to restore breathing in both sexes.
Collapse
|
41
|
Piechota M, Korostynski M, Golda S, Ficek J, Jantas D, Barbara Z, Przewlocki R. Transcriptional signatures of steroid hormones in the striatal neurons and astrocytes. BMC Neurosci 2017; 18:37. [PMID: 28381250 PMCID: PMC5381047 DOI: 10.1186/s12868-017-0352-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 03/08/2017] [Indexed: 01/05/2023] Open
Abstract
Background The mechanisms of steroids actions in the brain mainly involve the binding and nuclear translocation of specific cytoplasmic receptors. These receptors can act as transcription factors and regulate gene expression. However, steroid-dependent transcriptional regulation in different types of neural cells is not yet fully understood. The aim of this study was to evaluate and compare transcriptional alterations induced by various steroid receptor agonists in primary cultures of astrocytes and neurons from mouse brain. Results We utilized whole-genome microarrays (Illumina Mouse WG-6) and quantitative PCR analyses to measure mRNA abundance levels. To stimulate gene expression we treated neuronal and astroglial cultures with dexamethasone (100 nM), aldosterone (200 nM), progesterone (200 nM), 5α-dihydrotestosterone (200 nM) and β-Estradiol (200 nM) for 4 h. Neurons were found to exhibit higher levels of expression of mineralocorticoid receptor, progesterone receptor and estrogen receptor 2 than astrocytes. However, higher mRNA level of glucocorticoid receptor mRNA was observed in astrocytes. We identified 956 genes regulated by steroids. In astrocytes we found 381 genes altered by dexamethasone and 19 altered by aldosterone. Functional classification of the regulated genes indicated their putative involvement in multiple aspects of cell metabolism (up-regulated Slc2a1, Pdk4 and Slc45a3) and the inflammatory response (down-regulated Ccl3, Il1b and Tnf). Progesterone, dihydrotestosterone and estradiol did not change gene expression in astrocytes. We found no significant changes in gene expression in neurons. Conclusions The obtained results indicate that glial cells might be the primary targets of transcriptional action of steroids in the central nervous system. Substantial changes in gene expression driven by the glucocorticoid receptor imply an important role for the hypothalamic–pituitary–adrenal axis in the hormone-dependent regulation of brain physiology. This is an in vitro study. Hence, the model may not accurately reflect all the effects of steroids on gene expression in neurons in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s12868-017-0352-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marcin Piechota
- Department of Molecular Neuropharmacology, Institute of Pharmacology Polish Academy of Sciences, Smetna 12, 31-343, Kraków, Poland.
| | - Michał Korostynski
- Department of Molecular Neuropharmacology, Institute of Pharmacology Polish Academy of Sciences, Smetna 12, 31-343, Kraków, Poland
| | - Slawomir Golda
- Department of Molecular Neuropharmacology, Institute of Pharmacology Polish Academy of Sciences, Smetna 12, 31-343, Kraków, Poland
| | - Joanna Ficek
- Department of Molecular Neuropharmacology, Institute of Pharmacology Polish Academy of Sciences, Smetna 12, 31-343, Kraków, Poland
| | - Danuta Jantas
- Department of Neuroendocrinology, Institute of Pharmacology Polish Academy of Sciences, Smetna 12, 31-343, Kraków, Poland
| | - Ziolkowska Barbara
- Department of Molecular Neuropharmacology, Institute of Pharmacology Polish Academy of Sciences, Smetna 12, 31-343, Kraków, Poland
| | - Ryszard Przewlocki
- Department of Molecular Neuropharmacology, Institute of Pharmacology Polish Academy of Sciences, Smetna 12, 31-343, Kraków, Poland
| |
Collapse
|
42
|
Zhao Y, Yu Y, Zhang Y, He L, Qiu L, Zhao J, Liu M, Zhang J. Letrozole regulates actin cytoskeleton polymerization dynamics in a SRC-1 dependent manner in the hippocampus of mice. J Steroid Biochem Mol Biol 2017; 167:86-97. [PMID: 27866972 DOI: 10.1016/j.jsbmb.2016.11.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/24/2016] [Accepted: 11/16/2016] [Indexed: 12/21/2022]
Abstract
In the hippocampus, local estrogens (E2) derived from testosterone that is catalyzed by aromatase play important roles in the regulation of hippocampal neural plasticity, but the underlying mechanisms remain unclear. The actin cytoskeleton contributes greatly to hippocampal synaptic plasticity; however, whether it is regulated by local E2 and the related mechanisms remain to be elucidated. In this study, we first examined the postnatal developmental profiles of hippocampal aromatase and specific proteins responsible for actin cytoskeleton dynamics. Then we used aromatase inhibitor letrozole (LET) to block local E2 synthesis and examined the changes of these proteins and steroid receptor coactivator-1 (SRC-1), the predominant coactivator for steroid nuclear receptors. Finally, SRC-1 specific RNA interference was used to examine the effects of SRC-1 on the expression of these actin remodeling proteins. The results showed a V-type profile for aromatase and increased profiles for actin cytoskeleton proteins in both male and female hippocampus without obvious sex differences. LET treatment dramatically decreased the F-actin/G-actin ratio, the expression of Rictor, phospho-AKT (ser473), Profilin-1, phospho-Cofilin (Ser3), and SRC-1 in a dose-dependent manner. In vitro studies demonstrated that LET induced downregulation of these proteins could be reversed by E2, and E2 induced increase of these proteins were significantly suppressed by SRC-1 shRNA interference. These results for the first time clearly demonstrated that local E2 inhibition could induce aberrant actin polymerization; they also showed an important role of SRC-1 in the mediation of local E2 action on hippocampal synaptic plasticity by regulation of actin cytoskeleton dynamics.
Collapse
Affiliation(s)
- Yangang Zhao
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Yanlan Yu
- Student Brigade, Third Military Medical University, Chongqing 400038, China
| | - Yuanyuan Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Li He
- School of Nursing, Third Military Medical University, Chongqing 400038, China
| | - Linli Qiu
- School of Nursing, Third Military Medical University, Chongqing 400038, China; Department of Nursing, Sichuan Nursing Vocational College, Chengdu 610100, China
| | - Jikai Zhao
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Mengying Liu
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Jiqiang Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
43
|
Aoki C, Chowdhury TG, Wable GS, Chen YW. Synaptic changes in the hippocampus of adolescent female rodents associated with resilience to anxiety and suppression of food restriction-evoked hyperactivity in an animal model for anorexia nervosa. Brain Res 2017; 1654:102-115. [PMID: 26779892 PMCID: PMC4947030 DOI: 10.1016/j.brainres.2016.01.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/10/2016] [Indexed: 12/30/2022]
Abstract
Anorexia nervosa is a mental illness that emerges primarily during early adolescence, with mortality rate that is 200 times higher than that of suicide. The illness is characterized by intense fear of gaining weight, heightened anxiety, obstinate food restriction, often accompanied by excessive exercise, in spite of mounting hunger. The illness affects females nine times more often than males, suggesting an endocrine role in its etiology. Its relapse rate exceeds 25%, yet there are no accepted pharmacological treatments to prevent this. Here, we summarize studies from this laboratory that have used adolescent female rodents in activity-based anorexia (ABA), an animal model of anorexia nervosa, with the goal of identifying neurobiological underpinnings of this disease. We put forth a hypothesis that a GABAergic mechanism within the hippocampus is central to regulating an individual׳s anxiety which, in turn, strongly influences the individual׳s resilience/vulnerability to ABA. In particular, we propose that ionotropic GABAA receptors containing the subunits alpha4 and delta, are at play for exerting shunting inhibition upon hippocampal pyramidal neurons that become more excitable during ABA. Since these receptors confer insensitivity to benzodiazepines, this pharmacological profile of ABA fits with lack of report indicating efficacy of benzodiazepines in reducing the anxiety experienced by individuals with anorexia nervosa. The idea that the GABAergic system of the hippocampus regulates resilience/vulnerability to anorexia nervosa complements current opinions about the important roles of the prefrontal cortex, amygdala, striatum, gustatory pathways and feeding centers of the hypothalamus and of the neuromodulators, serotonin and dopamine, in the etiology of the disease. This article is part of a Special Issue entitled SI: Adolescent plasticity.
Collapse
Affiliation(s)
- Chiye Aoki
- Center for Neural Science, 4 Washington Place, Room 809, New York, NY 10003, United States.
| | - Tara G Chowdhury
- Center for Neural Science, 4 Washington Place, Room 809, New York, NY 10003, United States
| | - Gauri S Wable
- Center for Neural Science, 4 Washington Place, Room 809, New York, NY 10003, United States
| | - Yi-Wen Chen
- Center for Neural Science, 4 Washington Place, Room 809, New York, NY 10003, United States
| |
Collapse
|
44
|
Frange C, Banzoli CV, Colombo AE, Siegler M, Coelho G, Bezerra AG, Csermak M, Naufel MF, Cesar-Netto C, Andersen ML, Girão MJBC, Tufik S, Hachul H. Women's Sleep Disorders: Integrative Care. ACTA ACUST UNITED AC 2017; 10:174-180. [PMID: 29410750 PMCID: PMC5760052 DOI: 10.5935/1984-0063.20170030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The integrative care model is rooted in a biopsychosocial approach. Integrative
is a term which refers to increasing the harmony and coherence of your whole
being, and integrative care is therefore focused on the person, not on either
the disease or a therapy. It is provided collaboratively by a health team
comprising physicians, psychologists, physiotherapists, acupuncturists, and
meditation, nutrition, and floral therapy. Previous studies have demonstrated
that interventions based on the integrative care model improved womens lifestyle
and quality of life. Our aim was to describe the use of complementary and
alternative medicine (CAM) alongside traditional medicine among women with sleep
conditions in our Womens Sleep Disorders Integrative Treatment Outpatient
Clinic. We are sharing our experiences and clinical practice as the model we
developed seems to have both physical and psychological benefits for women with
sleep problems. We discuss the wide range of benefits that result from this type
of complex intervention, and the contextual factors that may influence these
benefits. This will inform future practitioners and we hope to contribute to
quantitative research in the clinical setting. The study highlights the
importance of treating sleep complaints with a caring relationship and a CAM
approach, alongside conventional medicine. Exploration of the lived experience
of CAM and its meaning enables healthcare professionals to gain insights into
the patients needs, preferences, and values. Gynecologists, clinicians, and
health care providers should support and guide patients in their decision to use
CAM by providing evidence-based and comprehensive advice on the potential
benefits, risks and related safety issues of this approach.
Collapse
Affiliation(s)
- Cristina Frange
- Universidade Federal de São Paulo, Departament Psychobiology - São Paulo - SP - Brazil
| | | | - Ana Elisa Colombo
- Universidade Federal de São Paulo, Department Gynecology - São Paulo - SP - Brazil
| | - Marcele Siegler
- Universidade Federal de São Paulo, Departament Psychobiology - São Paulo - SP - Brazil
| | - Glaury Coelho
- Universidade Federal de São Paulo, Departament Psychobiology - São Paulo - SP - Brazil
| | - Andréia Gomes Bezerra
- Universidade Federal de São Paulo, Departament Psychobiology - São Paulo - SP - Brazil
| | - Marcelo Csermak
- Universidade Federal de São Paulo, Departament Psychobiology - São Paulo - SP - Brazil
| | | | - Cristiana Cesar-Netto
- Universidade Federal de São Paulo, Departament Psychobiology - São Paulo - SP - Brazil
| | - Monica Levy Andersen
- Universidade Federal de São Paulo, Departament Psychobiology - São Paulo - SP - Brazil
| | | | - Sergio Tufik
- Universidade Federal de São Paulo, Departament Psychobiology - São Paulo - SP - Brazil
| | - Helena Hachul
- Universidade Federal de São Paulo, Departament Psychobiology - São Paulo - SP - Brazil.,Universidade Federal de São Paulo, Department Gynecology - São Paulo - SP - Brazil
| |
Collapse
|
45
|
Montelli S, Suman M, Corain L, Cozzi B, Peruffo A. Sexually Diergic Trophic Effects of Estradiol Exposure on Developing Bovine Cerebellar Granule Cells. Neuroendocrinology 2017; 104:51-71. [PMID: 26882349 DOI: 10.1159/000444528] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 02/06/2016] [Indexed: 11/19/2022]
Abstract
In the mammalian brain, the differentiation of neural cells and the developmental organization of the underlying circuitry are influenced by steroid hormones. The estrogen 17-β estradiol (E2) is one of the most potent regulators of neural growth during prenatal life, synthetized locally from steroid precursors including prenatal testicular testosterone. Estradiol promotes brain differentiation counting sexually dimorphic neural circuits by binding to the estrogen receptors, ER-α and ER-β. The cerebellum has been described as a site of estrogen action and a potentially sexually dimorphic area. The goal of this study was to analyze the capacity of E2 to affect the growth of male and female fetal bovine cerebellar granule. We performed primary cultures of fetal cerebellar granules, and verified the mRNA expression of the ER-α and ER-β in both sexes. Moreover, the distribution of ERs in the male and female cerebellar granules of the second fetal stage was characterized by immunohistochemistry. We measured morphological parameters in presence (or absence) of estradiol administration, focusing on the variations of the dendritic branching pattern of granule neurons. By using the nonparametric combination and permutation testing approach, we proposed a sophisticated multivariate statistical analysis to demonstrate that E2 induces multifarious and dimorphic changes in the granule cells. E2 exerts trophic effects in both female and male granules and this effect is stronger in female. Male granules treated with E2 became similar to female control granule. Bos taurus species has a long gestation and a large brain that offers an interesting alternative in comparative neuroscience.
Collapse
Affiliation(s)
- Stefano Montelli
- Department of Comparative Biomedicine and Food Science of the University of Padova, Legnaro, taly
| | | | | | | | | |
Collapse
|
46
|
Neurosteroids Involvement in the Epigenetic Control of Memory Formation and Storage. Neural Plast 2016; 2016:5985021. [PMID: 28090360 PMCID: PMC5206442 DOI: 10.1155/2016/5985021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 11/10/2016] [Indexed: 12/14/2022] Open
Abstract
Memory is our ability to store and remember past experiences; it is the result of changes in neuronal circuits of specific brain areas as the hippocampus. During memory formation, neurons integrate their functions and increase the strength of their connections, so that synaptic plasticity is improved and consolidated. All these processes recruit several proteins at the synapses, whose expression is highly regulated by DNA methylation and histone tails posttranslational modifications. Steroids are known to influence memory process, and, among them, neurosteroids are implicated in neurodegenerative disease related to memory loss and cognitive impairment. The epigenetic control of neurosteroids involvement in memory formation and maintenance could represent the basis for neuroregenerative therapies.
Collapse
|
47
|
Chen JR, Lim SH, Chung SC, Lee YF, Wang YJ, Tseng GF, Wang TJ. Reproductive experience modified dendritic spines on cortical pyramidal neurons to enhance sensory perception and spatial learning in rats. Exp Anim 2016; 66:61-74. [PMID: 27784858 PMCID: PMC5301002 DOI: 10.1538/expanim.16-0061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Behavioral adaptations during motherhood are aimed at increasing reproductive success.
Alterations of hormones during motherhood could trigger brain morphological changes to
underlie behavioral alterations. Here we investigated whether motherhood changes a rat’s
sensory perception and spatial memory in conjunction with cortical neuronal structural
changes. Female rats of different statuses, including virgin, pregnant, lactating, and
primiparous rats were studied. Behavioral test showed that the lactating rats were most
sensitive to heat, while rats with motherhood and reproduction experience outperformed
virgin rats in a water maze task. By intracellular dye injection and computer-assisted
3-dimensional reconstruction, the dendritic arbors and spines of the layer III and V
pyramidal neurons of the somatosensory cortex and CA1 hippocampal pyramidal neurons were
revealed for closer analysis. The results showed that motherhood and reproductive
experience increased dendritic spines but not arbors or the lengths of the layer III and V
pyramidal neurons of the somatosensory cortex and CA1 hippocampal pyramidal neurons. In
addition, lactating rats had a higher incidence of spines than pregnant or primiparous
rats. The increase of dendritic spines was coupled with increased expression of the
glutamatergic postsynaptic marker protein (PSD-95), especially in lactating rats. On the
basis of the present results, it is concluded that motherhood enhanced rat sensory
perception and spatial memory and was accompanied by increases in dendritic spines on
output neurons of the somatosensory cortex and CA1 hippocampus. The effect was sustained
for at least 6 weeks after the weaning of the pups.
Collapse
Affiliation(s)
- Jeng-Rung Chen
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Sleeping problems are a serious public health problem, imposing a substantial burden on individuals and society. Although sleeping problems occur throughout the lifespan, and in both sexes, menopause can be considered as one important milestone of increasing occurrence in sleeping problems. However, to determine whether sleeping problems are caused by the menopause or merely occur by coincidence during the menopause is not always easy because several, particularly age-related, changes take place at the same time. The most important factors are general diseases, medications, weight changes and mood symptoms. According to women's own judgment, hormone therapy significantly improves sleep quality. Hormone therapy can thus be considered as a first-line treatment for climacteric sleeping problems. If sleeping problems are accompanied by other disorders, hormone therapy should be kept in mind as an adjuvant therapy. According to worldwide consensus on hormone therapy, the main indication of hormone therapy is alleviation of climacteric symptoms, including climacteric sleeping problems. However, when choosing hormone therapy for a patient, contraindications and possible long-term side effects should be individually considered. This review illustrates the effect of menopause on sleep and evaluates different treatment options, especially hormone therapy, in alleviation of symptoms.
Collapse
Affiliation(s)
- Päivi Polo-Kantola
- University of Turku, Department of Obstetrics and Gynecology, University Central Hospital of Turku and Sleep Research Center Dentalia, FIN-20520 Turku, Finland, Tel.: +358 2313 0000; Fax: +358 2313 2340
| |
Collapse
|
49
|
Shua-Haim JR, Ross JS. Current and the near future medications for Alzheimer's disease: What can we expect from them? ACTA ACUST UNITED AC 2016. [DOI: 10.1177/153331759901400507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
| | - J. S. Ross
- Outpatient Geriatric Department, Jersey Shore Medical Center, The Medical Center of Ocean County, Meridian Health System, and the Center of Aging at UMDNJ SOM, Stratford, New Jersey
| |
Collapse
|
50
|
Glynn LM, Davis EP, Sandman CA, Goldberg WA. Gestational hormone profiles predict human maternal behavior at 1-year postpartum. Horm Behav 2016; 85:19-25. [PMID: 27427279 PMCID: PMC5929113 DOI: 10.1016/j.yhbeh.2016.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 06/17/2016] [Accepted: 07/13/2016] [Indexed: 12/26/2022]
Abstract
In many non-human species, including primates, gestational reproductive hormones play an essential role in the onset of maternal motivation and behaviors. We investigated the associations between prepartum estradiol and progesterone and maternal behavior at 1-year postpartum in 177 women. Blood was obtained at five gestational time points and an index of quality of maternal care was determined using a well-validated mother-child interaction protocol. Women who exhibited higher quality maternal care at 1-year postpartum were characterized by unique gestational profiles of estradiol, progesterone and the estrogen to progesterone ratio; specifically by slower accelerations and levels of these hormone trajectories beginning in midgestation. Further, it appeared that both fetal sex and parity moderated these findings, with first time mothers and mothers of females showing stronger associations. In sum, these data document persisting associations between prepartum hormone profiles and human maternal behavior. More broadly, these findings add to the growing literature highlighting the perinatal period as one of critical neurodevelopment in the lifespan of the human female.
Collapse
Affiliation(s)
- Laura M Glynn
- Department of Psychology, Chapman University, One University Dr., Orange, CA 92868, United States; Department of Psychiatry & Human Behavior, University of California, Irvine, 101 The City Dr., Building 3, Route 88, Orange, CA 92868, United States.
| | - Elysia Poggi Davis
- Department of Psychiatry & Human Behavior, University of California, Irvine, 101 The City Dr., Building 3, Route 88, Orange, CA 92868, United States; Department of Psychology, University of Denver, 2155 South Race St., Denver, CO 80210, United States
| | - Curt A Sandman
- Department of Psychiatry & Human Behavior, University of California, Irvine, 101 The City Dr., Building 3, Route 88, Orange, CA 92868, United States
| | - Wendy A Goldberg
- Department of Psychology and Social Behavior, University of California, Irvine, 5300 Social and Behavioral Sciences Gateway, Irvine, CA 92697, United States
| |
Collapse
|