1
|
Flannery JC, Tirrell PS, Baumgartner NE, Daniel JM. Neuroestrogens, the hippocampus, and female cognitive aging. Horm Behav 2025; 170:105710. [PMID: 40036999 DOI: 10.1016/j.yhbeh.2025.105710] [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: 08/30/2024] [Revised: 12/30/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
Research conducted over the last several decades implicates ovarian estrogens as important modulators of hippocampal function. More recently however, the importance of estrogens synthesized in the brain de novo for hippocampal function has been recognized. These brain-derived neuroestrogens act in the hippocampus to regulate dendritic spine dynamics and synaptic plasticity as well as hippocampus-dependent memory. The current report provides an overview of research conducted in model systems elucidating the actions of neuroestrogens in the hippocampus and the subsequent consequences for cognition. We highlight the relationship between ovarian estrogens and brain-derived estrogens and discuss implications for female cognitive aging of the putative decline in hippocampal levels of neuroestrogens following loss of ovarian function. Finally, we propose a model of menopause in which a short-term period of midlife estradiol treatment changes the trajectory of hippocampal neuroestrogen production long-term, resulting in sustained interactions of neuroestrogens, insulin-like growth factor-1, and estrogen receptor signaling in the hippocampus, interactions that support successful brain and cognitive aging.
Collapse
Affiliation(s)
- Jill C Flannery
- Brain Institute, Tulane University, New Orleans, LA 70118, United States of America; Neuroscience Program, Tulane University, New Orleans, LA 70118, United States of America
| | - Parker S Tirrell
- Brain Institute, Tulane University, New Orleans, LA 70118, United States of America; Neuroscience Program, Tulane University, New Orleans, LA 70118, United States of America
| | - Nina E Baumgartner
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, United States of America
| | - Jill M Daniel
- Brain Institute, Tulane University, New Orleans, LA 70118, United States of America; Neuroscience Program, Tulane University, New Orleans, LA 70118, United States of America; Department of Psychology, Tulane University, New Orleans, LA, 70118, United States of America.
| |
Collapse
|
2
|
Fischer L, Paschke B, Gareis F, Schumacher M, Liere P, Hiergeist A, Gessner A, Rupprecht R, Neumann ID, Bosch OJ. The translocator protein 18 kDa (TSPO) ligand etifoxine in an animal model of anxiety: Line- and sex-dependent effects on emotionality, stress reactivity, spine density, oxytocin receptors, steroids, and microbiome composition. Neuropharmacology 2025; 266:110282. [PMID: 39725124 DOI: 10.1016/j.neuropharm.2024.110282] [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: 11/02/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
The treatment of stress-related disorders such as anxiety and depression is still challenging. One potential therapeutical option are neurosteroids. Their synthesis is promoted by ligands of the mitochondrial translocator protein 18 kDa (TSPO). We tested the TSPO ligand etifoxine (ETX) in a rat model of hyper-anxiety and depression-like behavior, i.e., in female and male HAB (high anxiety-related behavior) rats, as well as in respective low anxiety (LAB) and non-selected control (NAB) rats for behavioral, molecular, cellular, and physiological parameters. Daily acute i.p. treatment with ETX or vehicle over 5 or 9 days revealed that ETX was most effective in female HAB rats; it reduced anxiety levels (5 days) and OXT-R binding brain site-specifically (5 and 9 days), and increased spine density (5 days). The behavioral ETX effect exclusively found in female HABs was accompanied by increased 3β5α-THDOC levels, without any effect in female LABs and NABs and on other neurosteroids. In males of all breeding lines, ETX changed a total of 10 out of 23 brain steroids. Passive stress-coping during 10-min forced swimming was not affected by 9-day treatment with ETX, the resulting stress-induced plasma corticosterone levels were higher in ETX-treated NAB rats of both sexes compared with their VEH-treated groups. The fecal bacterial composition was similar but beta diversity differed between HABs and LABs and from NABs independent of sex; ETX treatment had no effect. Therefore, we propose considering the aspect of sex in treatment strategies for anxiety disorders. This is particularly important to establish better treatment regimens for women.
Collapse
Affiliation(s)
- Lilith Fischer
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| | - Bjarne Paschke
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| | - Franziska Gareis
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| | - Michael Schumacher
- U1195 Inserm and University Paris-Saclay, 80 Rue Du Général Leclerc, Le Kremlin-Bicêtre, 94276, France.
| | - Philippe Liere
- U1195 Inserm and University Paris-Saclay, 80 Rue Du Général Leclerc, Le Kremlin-Bicêtre, 94276, France.
| | - Andreas Hiergeist
- Institute of Clinical Microbiology and Hygiene, University Medical Center, 93053 Regensburg, Germany.
| | - André Gessner
- Institute of Clinical Microbiology and Hygiene, University Medical Center, 93053 Regensburg, Germany.
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany.
| | - Inga D Neumann
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| | - Oliver J Bosch
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
3
|
Aspesi D, Bass N, Kavaliers M, Choleris E. The Role of Androgens and Estrogens in Social Interactions and Social Cognition. Neuroscience 2025; 568:476-502. [PMID: 37080448 DOI: 10.1016/j.neuroscience.2023.03.028] [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: 06/13/2022] [Revised: 03/02/2023] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
Gonadal hormones are becoming increasingly recognized for their effects on cognition. Estrogens, in particular, have received attention for their effects on learning and memory that rely upon the functioning of various brain regions. However, the impacts of androgens on cognition are relatively under investigated. Testosterone, as well as estrogens, have been shown to play a role in the modulation of different aspects of social cognition. This review explores the impact of testosterone and other androgens on various facets of social cognition including social recognition, social learning, social approach/avoidance, and aggression. We highlight the relevance of considering not only the actions of the most commonly studied steroids (i.e., testosterone, 17β-estradiol, and dihydrotestosterone), but also that of their metabolites and precursors, which interact with a plethora of different receptors and signalling molecules, ultimately modulating behaviour. We point out that it is also essential to investigate the effects of androgens, their precursors and metabolites in females, as prior studies have mostly focused on males. Overall, a comprehensive analysis of the impact of steroids such as androgens on behaviour is fundamental for a full understanding of the neural mechanisms underlying social cognition, including that of humans.
Collapse
Affiliation(s)
- Dario Aspesi
- Department of Psychology and Neuroscience Program, University of Guelph, Canada
| | - Noah Bass
- Department of Psychology and Neuroscience Program, University of Guelph, Canada
| | - Martin Kavaliers
- Department of Psychology and Neuroscience Program, University of Guelph, Canada; Department of Psychology, University of Western Ontario, London, Canada; Graduate Program in Neuroscience, University of Western Ontario, London, Canada
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Canada.
| |
Collapse
|
4
|
Duarte-Guterman P, Skandalis DA, Merkl A, Geissler DB, Ehret G. Brain aromatase and its relationship with parental experience and behavior in male mice. Front Neurosci 2025; 19:1502764. [PMID: 40035063 PMCID: PMC11872740 DOI: 10.3389/fnins.2025.1502764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction In most mammals, paternal care is not mandatory for raising offspring. In house mice, experience with pups governs the extent and quality of paternal care. First-time fathers undergo a dramatic transition from ignoring or killing pups to caring for pups. The behavioral shift occurs together with changes in brain estrogen signaling as indicated by changes in estrogen receptor presence and distribution in multiple areas regulating olfaction, emotion, and motivation. Methods We measured changes in the expression of aromatase, the enzyme converting testosterone into estrogen, as an indirect measure of estrogen synthesis, in various areas of the limbic system in mice with increasing paternal experience. Results The amount of paternal experience (5 or 27 days) was associated with increased numbers of immunocytochemically-identified aromatase expressing cells in the medial and cortical amygdala, posterior piriform cortex, and ventromedial hypothalamus. Functionally, these changes can be related to the disappearance of aggression or neglect towards pups when first-time fathers or, even more, well-experienced fathers are handling their own pups. In the lateral septum, the anterior piriform cortex and to some extent in the medial preoptic area, parental experience increased the number of aromatase-positive cells only in fathers with 27 days of experience, and only in the right hemisphere. This represents a novel case of brain-functional lateralization triggered by experience. Nuclei/areas associated with maternal care (medial preoptic area, bed nucleus of stria terminalis, nucleus accumbens) exhibited a left-hemisphere advantage in aromatase expressing cells, both in pup-naïve and pup-experienced males. This newly found lateralization may contribute to the left-hemisphere dominant processing and perception of pup calls to release parental behavior. Conclusion In general, the experience-dependent changes in aromatase expression we observed in most brain areas did not mirror the previously reported changes in estrogen receptors (ERα) when pup-naïve males became pup-caring fathers. Hence, paternal behavior may depend, in a brain area-specific way, on the differential action of estrogen through its receptors and/or direct local modulation of neural processing.
Collapse
Affiliation(s)
| | | | - Ariane Merkl
- Institute of Neurobiology, University of Ulm, Ulm, Germany
| | | | - Günter Ehret
- Institute of Neurobiology, University of Ulm, Ulm, Germany
| |
Collapse
|
5
|
Bourque M, Morissette M, Isenbrandt A, Giatti S, Melcangi RC, Carta M, Frau R, Bortolato M, Soulet D, Di Paolo T. Effect of 5-alpha reductase inhibitors in animal models of Parkinson's disease. Front Neuroendocrinol 2024; 75:101156. [PMID: 39353534 DOI: 10.1016/j.yfrne.2024.101156] [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: 06/03/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Parkinson's disease (PD) is characterized by motor symptoms due to loss of brain dopamine and non-motor symptoms, including gastrointestinal disorders. Although there is no cure for PD, symptomatic treatments are available. L-Dopa is the gold standard PD therapy, but most patients develop dyskinesias (LID), which are challenging to manage. Amantadine is recognized as the most effective drug for LID, but its adverse effects limit the use in patients. Here we review how 5α-reductase inhibitors (5ARIs), drugs used to treat benign prostatic hyperplasia and alopecia, exhibit beneficial effects in PD animal models. 5ARIs show neuroprotective properties in brain and gut dopaminergic systems, and reduce dyskinesias in rodent model of PD. Additionally, the 5ARI finasteride dampened dopaminergic-induced drug gambling in PD patients. Neuroprotection and antidyskinetic activities of 5ARIs in animal models of PD suggest their potential repurposing in men with PD to address gut dysfunction, protect brain DA and inhibit dyskinesias.
Collapse
Affiliation(s)
- Mélanie Bourque
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Marc Morissette
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Amandine Isenbrandt
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada
| | - Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Manolo Carta
- Department of Biomedical Sciences, "Guy Everett Laboratory", University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, "Guy Everett Laboratory", University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT 84112, USA
| | - Denis Soulet
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada
| | - Thérèse Di Paolo
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada.
| |
Collapse
|
6
|
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
|
7
|
Melcangi RC. Role of Neuroactive Steroids in Health and Disease. Biomolecules 2024; 14:941. [PMID: 39199329 PMCID: PMC11352212 DOI: 10.3390/biom14080941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 09/01/2024] Open
Abstract
Steroidogenesis occurs not only in endocrine peripheral glands (i [...].
Collapse
Affiliation(s)
- Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
8
|
Barrón-González M, Rivera-Antonio AM, Jarillo-Luna RA, Santiago-Quintana JM, Levaro-Loquio D, Pérez-Capistran T, Guerra-Araiza CH, Soriano-Ursúa MA, Farfán-García ED. Borolatonin limits cognitive deficit and neuron loss while increasing proBDNF in ovariectomised rats. Fundam Clin Pharmacol 2024; 38:730-741. [PMID: 38423984 DOI: 10.1111/fcp.12997] [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: 11/21/2023] [Revised: 01/31/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Borolatonin is a potential therapeutic agent for some neuronal diseases such as Alzheimer's disease (AD). Its administration exerts ameliorative effects such as those induced by the equimolar administration of melatonin in behavioral tests on male rats and in neuronal immunohistochemistry assays. OBJECTIVE In this study, motivated by sex differences in neurobiology and the incidence of AD, the ability of borolatonin to induce changes in female rats was assessed. METHODS Effects of borolatonin were measured by the evaluation of both behavioral and immunohistopathologic approaches; additionally, its ability to limit amyloid toxicity was determined in vitro. RESULTS Surprisingly, behavioral changes were similar to those reported in male rats, but not those evaluated by immunoassays regarding neuronal survival; while pro-brain-derived neurotrophic factor (BDNF) immunoreactivity and the limitation of toxicity by amyloid in vitro were observed for the first time. CONCLUSION Borolatonin administration induced changes in female rats. Differences induced by the administration of borolatonin or melatonin could be related to the differences in the production of steroid hormones in sex dependence. Further studies are required to clarify the possible mechanism and origin of differences in disturbed memory caused by the gonadectomy procedure between male and female rats.
Collapse
Affiliation(s)
- Mónica Barrón-González
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Astrid M Rivera-Antonio
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, ESM-IPN, Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Rosa A Jarillo-Luna
- Laboratorio de Morfología, Sección de Estudios de Posgrado e Investigación, ESM-IPN, Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - José M Santiago-Quintana
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - David Levaro-Loquio
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Teresa Pérez-Capistran
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Christian H Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, Mexico
| | - Marvin A Soriano-Ursúa
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Eunice D Farfán-García
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| |
Collapse
|
9
|
Maddern XJ, Ursich LT, Bailey G, Pearl A, Anversa RG, Lawrence AJ, Walker LC. Sex Differences in Alcohol Use: Is It All About Hormones? Endocrinology 2024; 165:bqae088. [PMID: 39018449 DOI: 10.1210/endocr/bqae088] [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: 04/02/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/19/2024]
Abstract
Risky alcohol use and alcohol use disorders (AUD) are a rising problem in women, yet a major disparity in our understanding of what drives alcohol consumption in women remains. Historically biomedical research has focused on male subjects; however, recent increases in reporting of females, have highlighted major differences between the sexes. Here we review the current literature of the effect of gonadal steroid hormones (estrogens, androgens, and progestins), neurosteriods, and neurobiological factors on alcohol use in clinical and preclinical studies of both sexes. Further, we briefly discuss how fundamental sex differences in genetics, metabolism, neuroimmune, and stress responses may influence sex differences in alcohol intake. Comparing the sexes could aid in the discovery of novel therapeutics to treat AUD, and implementation of current treatment options in women.
Collapse
Affiliation(s)
- Xavier J Maddern
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Lauren T Ursich
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Grace Bailey
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Amy Pearl
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
| | - Roberta G Anversa
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| |
Collapse
|
10
|
Cioffi L, Grassi D, Diviccaro S, Caruso D, Pinto-Benito D, Arevalo MA, Garcia-Segura LM, Melcangi RC, Giatti S. Sex chromosome complement interacts with gonadal hormones in determining regional-specific neuroactive steroid levels in plasma, hippocampus, and hypothalamus. A study using the four core genotype mouse model. J Steroid Biochem Mol Biol 2024; 241:106514. [PMID: 38554982 DOI: 10.1016/j.jsbmb.2024.106514] [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: 02/02/2024] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
An important aspect of the neuromodulatory and neuroprotective actions exerted by neuroactive steroids is that they are sex-specific, as determined by the sexually dimorphic levels of these molecules in plasma and the nervous tissue. Thus, the identification of the factors that generate the sex-dimorphic levels of neuroactive steroids may be crucial from a neuroprotectant perspective. The main driver for sex determination in mammals is the SRY gene and the subsequent presence of a specific gonad: testes for males and ovaries for females, thus producing hormonal compounds, primarily androgens and estrogens, respectively. Nowadays, it is well established that despite the relevance of gonads, other factors control sexual features, and, among them, sex chromosome complement is highly relevant. In this study, neuroactive steroids were evaluated by liquid chromatography-tandem mass spectrometry in the hypothalamus, the hippocampus, and plasma of the four core genotype mouse model, to determine the relative contribution of sex chromosome complement and gonads in determining their sex dimorphic levels. The data obtained reveal that although gonads are the main contributing factor for sex differences in neuroactive steroid levels, the levels of some neuroactive steroids, including testosterone, are also influenced in brain and plasma by tissue-specific actions of sex chromosomes. The data presented here adds a new piece to the puzzle of steroid level regulation, which may be useful in designing sex-specific neuroprotective approaches to pathological conditions affecting the nervous system.
Collapse
Affiliation(s)
- Lucia Cioffi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Itlay
| | - Daniela Grassi
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Autonoma University of Madrid, Calle Arzobispo Morcillo 4, Madrid 28029, Spain
| | - Silvia Diviccaro
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Itlay
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Itlay
| | - Daniel Pinto-Benito
- 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
| | - Maria-Angeles Arevalo
- 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
| | - 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, Itlay
| | - Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milano 20133, Itlay
| |
Collapse
|
11
|
Hidalgo-Lanussa O, González Santos J, Barreto GE. Sex-specific vulnerabilities in human astrocytes underpin the differential impact of palmitic acid. Neurobiol Dis 2024; 195:106489. [PMID: 38552721 DOI: 10.1016/j.nbd.2024.106489] [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: 02/21/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024] Open
Abstract
Obesity and neurometabolic diseases have been linked to neurodegenerative diseases. Our hypothesis is that the endogenous estrogenic component of human astrocytes plays a critical role in cell response during lipotoxic damage, given that obesity can disrupt hormonal homeostasis and cause brain inflammation. Our findings showed that high concentrations of palmitic acid (PA) significantly reduced cell viability more in male astrocytes, indicating sex-specific vulnerabilities. PA induced a greater increase in cytosolic reactive oxygen species (ROS) production in males, while female astrocytes exhibited higher superoxide ion levels in mitochondria. In addition, female astrocytes treated with PA showed increased expression of antioxidant proteins, including catalase, Gpx-1 and Nrf2 suggesting a stronger cellular defence mechanism. Interestingly, there was a difference in the expression of estrogenic components, such as estrogen, androgens, and progesterone receptors, as well as aromatase and 5α-reductase enzymes, between males and females. PA induced their expression mainly in females, indicating a potential protective mechanism mediated by endogenous hormones. In summary, our findings highlight the impact of sex on the response of human astrocytes to lipotoxicity. Male astrocytes appear to be more susceptible to cellular damage when exposed to high concentrations of fatty acids.
Collapse
Affiliation(s)
- Oscar Hidalgo-Lanussa
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia; Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Janneth González Santos
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
| |
Collapse
|
12
|
Ågmo A. Androgen receptors and sociosexual behaviors in mammals: The limits of generalization. Neurosci Biobehav Rev 2024; 157:105530. [PMID: 38176634 DOI: 10.1016/j.neubiorev.2023.105530] [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: 10/18/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024]
Abstract
Circulating testosterone is easily aromatized to estradiol and reduced to dihydrotestosterone in target tissues and elsewhere in the body. Thus, the actions of testosterone can be mediated either by the estrogen receptors, the androgen receptor or by simultaneous action at both receptors. To determine the role of androgens acting at the androgen receptor, we need to eliminate actions at the estrogen receptors. Alternatively, actions at the androgen receptor itself can be eliminated. In the present review, I will analyze the specific role of androgen receptors in male and female sexual behavior as well as in aggression. Some comments about androgen receptors and social recognition are also made. It will be shown that there are important differences between species, even between strains within a species, concerning the actions of the androgen receptor on the behaviors mentioned. This fact makes generalizations from one species to another or from one strain to another very risky. The existence of important species differences is often ignored, leading to many misunderstandings and much confusion.
Collapse
Affiliation(s)
- Anders Ågmo
- Department of Psychology, University of Tromsø, Norway.
| |
Collapse
|
13
|
Knyzeliene A, Wimberley C, MacAskill MG, Alcaide-Corral CJ, Morgan TEF, Henry MC, Lucatelli C, Pimlott SL, Sutherland A, Tavares AAS. Sexually dimorphic murine brain uptake of the 18 kDa translocator protein PET radiotracer [ 18F]LW223. Brain Commun 2024; 6:fcae008. [PMID: 38304004 PMCID: PMC10833650 DOI: 10.1093/braincomms/fcae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/09/2023] [Accepted: 01/12/2024] [Indexed: 02/03/2024] Open
Abstract
The 18 kDa translocator protein is a well-known biomarker of neuroinflammation, but also plays a role in homeostasis. PET with 18 kDa translocator protein radiotracers [11C]PBR28 in humans and [18F]GE180 in mice has demonstrated sex-dependent uptake patterns in the healthy brain, suggesting sex-dependent 18 kDa translocator protein expression, although humans and mice had differing results. This study aimed to assess whether the 18 kDa translocator protein PET radiotracer [18F]LW223 exhibited sexually dimorphic uptake in healthy murine brain and peripheral organs. Male and female C57Bl6/J mice (13.6 ± 5.4 weeks, 26.8 ± 5.4 g, mean ± SD) underwent 2 h PET scanning post-administration of [18F]LW223 (6.7 ± 3.6 MBq). Volume of interest and parametric analyses were performed using standard uptake values (90-120 min). Statistical differences were assessed by unpaired t-test or two-way ANOVA with Šidak's test (alpha = 0.05). The uptake of [18F]LW223 was significantly higher across multiple regions of the male mouse brain, with the most pronounced difference detected in hypothalamus (P < 0.0001). Males also exhibited significantly higher [18F]LW223 uptake in the heart when compared to females (P = 0.0107). Data support previous findings on sexually dimorphic 18 kDa translocator protein radiotracer uptake patterns in mice and highlight the need to conduct sex-controlled comparisons in 18 kDa translocator protein PET imaging studies.
Collapse
Affiliation(s)
- Agne Knyzeliene
- British Heart Foundation-University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Catriona Wimberley
- Edinburgh Imaging, University of Edinburgh, Edinburgh EH16 4TJ, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Mark G MacAskill
- British Heart Foundation-University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Carlos J Alcaide-Corral
- British Heart Foundation-University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Timaeus E F Morgan
- British Heart Foundation-University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Martyn C Henry
- School of Chemistry, University of Glasgow, Glasgow G12 8QQ, UK
| | | | - Sally L Pimlott
- West of Scotland PET Centre, Greater Glasgow and Clyde NHS Trust, Glasgow G12 0YN, UK
| | | | - Adriana A S Tavares
- British Heart Foundation-University of Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh EH16 4TJ, UK
| |
Collapse
|
14
|
Lawande NV, Conklin EA, Christian‐Hinman CA. Sex and gonadectomy modify behavioral seizure susceptibility and mortality in a repeated low-dose kainic acid systemic injection paradigm in mice. Epilepsia Open 2023; 8:1512-1522. [PMID: 37715318 PMCID: PMC10690657 DOI: 10.1002/epi4.12828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023] Open
Abstract
OBJECTIVE Sex differences in epilepsy appear driven in part due to effects of gonadal steroids, with varying results in experimental models based on species, strain, and method of seizure induction. Furthermore, removing the main source of these steroids via gonadectomy may impact seizure characteristics differently in males and females. Repeated low-dose kainic acid (RLDKA) systemic injection paradigms were recently shown to reliably induce status epilepticus (SE) and hippocampal histopathology in C57BL/6J mice. Here, we investigated whether seizure susceptibility in a RLDKA injection protocol exhibits a sex difference and whether gonadectomy differentially influences response to this seizure induction paradigm in males and females. METHODS Adult C57BL/6J mice were left gonad-intact as controls or gonadectomized (females: ovariectomized, OVX; males: orchidectomized, ORX). At least 2 weeks later, KA was injected ip, every 30 minutes at 7.5 mg/kg or less until the animal reached SE, defined by at least 5 generalized seizures (GS, Racine stage 3 or higher). Parameters of susceptibility to GS induction, SE development, and mortality rates were quantified. RESULTS No differences in seizure susceptibility or mortality were observed between control males and control females. Gonadectomized mice exhibited increased susceptibility and reduced latency to both GS and SE in comparison to corresponding controls of the same sex, but the effects were stronger in males. In addition, ORX males, but not OVX females, exhibited strongly increased seizure-induced mortality. SIGNIFICANCE The RLDKA protocol is notable for its efficacy in inducing SE and seizure-induced histopathology in C57BL/6J mice, the background for many transgenic strains in current use in epilepsy research. The present results indicate that this protocol may be beneficial for investigating the effects of gonadal hormone replacement on seizure susceptibility, mortality, and seizure-induced histopathology, and that gonadectomy unmasks sex differences in susceptibility to seizures and mortality not observed in gonad-intact controls.
Collapse
Affiliation(s)
- Niraj V. Lawande
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | | | - Catherine A. Christian‐Hinman
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Neuroscience ProgramUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
15
|
Seib DR, Tobiansky DJ, Meitzen J, Floresco SB, Soma KK. Neurosteroids and the mesocorticolimbic system. Neurosci Biobehav Rev 2023; 153:105356. [PMID: 37567491 PMCID: PMC11915106 DOI: 10.1016/j.neubiorev.2023.105356] [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: 12/29/2022] [Revised: 08/01/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
The mesocorticolimbic system coordinates executive functions, such as working memory and behavioral flexibility. This circuit includes dopaminergic projections from the ventral tegmental area to the nucleus accumbens and medial prefrontal cortex. In this review, we summarize evidence that cells in multiple nodes of the mesocorticolimbic system produce neurosteroids (steroids synthesized in the nervous system) and express steroid receptors. Here, we focus on neuroandrogens (androgens synthesized in the nervous system), neuroestrogens (estrogens synthesized in the nervous system), and androgen and estrogen receptors. We also summarize how (neuro)androgens and (neuro)estrogens affect dopamine signaling in the mesocorticolimbic system and regulate executive functions. Taken together, the data suggest that steroids produced in the gonads and locally in the brain modulate higher-order cognition and executive functions.
Collapse
Affiliation(s)
- Désirée R Seib
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| | - Daniel J Tobiansky
- Department of Biology and Neuroscience Program, St. Mary's College of Maryland, St. Mary's City, MD, USA
| | - John Meitzen
- Department of Biological Sciences and Center for Human Health and the Environment, NC State University, Raleigh, NC, USA
| | - Stan B Floresco
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Kiran K Soma
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
16
|
Diviccaro S, Cioffi L, Piazza R, Caruso D, Melcangi RC, Giatti S. Neuroactive Steroid-Gut Microbiota Interaction in T2DM Diabetic Encephalopathy. Biomolecules 2023; 13:1325. [PMID: 37759725 PMCID: PMC10527303 DOI: 10.3390/biom13091325] [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: 07/12/2023] [Revised: 08/11/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
The pathological consequences of type 2 diabetes mellitus (T2DM) also involve the central nervous system; indeed, T2DM patients suffer from learning and memory disabilities with a higher risk of developing dementia. Although several factors have been proposed as possible contributors, how neuroactive steroids and the gut microbiome impact brain pathophysiology in T2DM remain unexplored. On this basis, in male Zucker diabetic fatty (ZDF) rats, we studied whether T2DM alters memory abilities using the novel object recognition test, neuroactive steroid levels by liquid chromatography-tandem mass spectrometry, hippocampal parameters using molecular assessments, and gut microbiome composition using 16S next-generation sequencing. Results obtained reveal that T2DM worsens memory abilities and that these are correlated with increased levels of corticosterone in plasma and with a decrease in allopregnanolone in the hippocampus, where neuroinflammation, oxidative stress, and mitochondrial dysfunction were reported. Interestingly, our analysis highlighted a small group of taxa strictly related to both memory impairment and neuroactive steroid levels. Overall, the data underline an interesting role for allopregnanolone and microbiota that may represent candidates for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (S.D.); (L.C.); (D.C.); (R.C.M.)
| | - Lucia Cioffi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (S.D.); (L.C.); (D.C.); (R.C.M.)
| | - Rocco Piazza
- Dipartimento di Medicina e Chirurgia, Università di Milano—Bicocca, 20126 Milan, Italy;
| | - Donatella Caruso
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (S.D.); (L.C.); (D.C.); (R.C.M.)
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (S.D.); (L.C.); (D.C.); (R.C.M.)
| | - Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (S.D.); (L.C.); (D.C.); (R.C.M.)
| |
Collapse
|
17
|
Lawande NV, Conklin EA, Christian-Hinman CA. Sex and gonadectomy modify behavioral seizure susceptibility and mortality in a repeated low-dose kainic acid systemic injection paradigm in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541824. [PMID: 37292790 PMCID: PMC10245840 DOI: 10.1101/2023.05.22.541824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Objective Sex differences in epilepsy appear driven in part due to effects of gonadal steroids, with varying results in experimental models based on species, strain, and method of seizure induction. Furthermore, removing a main source of these steroids via gonadectomy may impact seizure characteristics differently in males and females. Repeated low-dose kainic acid (RLDKA) systemic injection paradigms were recently shown to reliably induce status epilepticus (SE) and hippocampal histopathology in C57BL/6J mice. Here, we investigated whether seizure susceptibility in a RLDKA injection protocol exhibits a sex difference, and whether gonadectomy differentially influences response to this seizure induction paradigm in males and females. Methods Adult C57BL/6J mice were left gonad-intact as controls or gonadectomized (females: ovariectomized, OVX; males: orchidectomized, ORX). At least 2 weeks later, KA was injected i.p. every 30 minutes at 7.5 mg/kg or less until the animal reached SE, defined by at least 5 generalized seizures (GS, Racine stage 3 or higher). Parameters of susceptibility to GS induction, SE development, and mortality rates were quantified. Results No differences in seizure susceptibility or mortality were observed between control males and control females. ORX males exhibited increased susceptibility and reduced latency to both GS and SE, but OVX females exhibited increased susceptibility and reduced latency to SE only. However, ORX males, but not OVX females, exhibited strongly increased seizure-induced mortality. Significance The RLDKA protocol is notable for its efficacy in inducing SE and seizure-induced histopathology in C57BL/6J mice, the background for many transgenic strains in current use in epilepsy research. The present results indicate that this protocol may be beneficial for investigating the effects of gonadal hormone replacement on seizure susceptibility, mortality, and seizure-induced histopathology, and that gonadectomy unmasks sex differences in susceptibility to seizures and mortality not observed in gonad-intact controls.
Collapse
Affiliation(s)
- Niraj V. Lawande
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
| | - Elisabeth A. Conklin
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
| | - Catherine A. Christian-Hinman
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
| |
Collapse
|
18
|
Diabetic Encephalopathy in a Preclinical Experimental Model of Type 1 Diabetes Mellitus: Observations in Adult Female Rat. Int J Mol Sci 2023; 24:ijms24021196. [PMID: 36674713 PMCID: PMC9860834 DOI: 10.3390/ijms24021196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 01/11/2023] Open
Abstract
Patients affected by diabetes mellitus (DM) show diabetic encephalopathy with an increased risk of cognitive deficits, dementia and Alzheimer's disease, but the mechanisms are not fully explored. In the male animal models of DM, the development of cognitive impairment seems to be the result of the concomitance of different processes such as neuroinflammation, oxidative stress, mitochondrial dysfunction, and aberrant synaptogenesis. However, even if diabetic encephalopathy shows some sex-dimorphic features, no observations in female rats have been so far reported on these aspects. Therefore, in an experimental model of type 1 DM (T1DM), we explored the impact of one month of pathology on memory abilities by the novel object recognition test and on neuroinflammation, synaptogenesis and mitochondrial functionality. Moreover, given that steroids are involved in memory and learning, we also analysed their levels and receptors. We reported that memory dysfunction can be associated with different features in the female hippocampus and cerebral cortex. Indeed, in the hippocampus, we observed aberrant synaptogenesis and neuroinflammation but not mitochondrial dysfunction and oxidative stress, possibly due to the results of locally increased levels of progesterone metabolites (i.e., dihydroprogesterone and allopregnanolone). These observations suggest specific brain-area effects of T1DM since different alterations are observed in the cerebral cortex.
Collapse
|
19
|
Tongta S, Daendee S, Kalandakanond-Thongsong S. Anxiety-like behavior and GABAergic system in ovariectomized rats exposed to chronic mild stress. Physiol Behav 2023; 258:114014. [PMID: 36328075 DOI: 10.1016/j.physbeh.2022.114014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/14/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
Stress or low level of estrogen could promote anxiety and depression; thus, it is of interest to investigate the combined effect of mild stress and the lack of estrogen on mental disorders by utilizing an animal model. This study was conducted to assess anxiety- and depressive- like behaviors in ovariectomized (Ovx) rats exposed to chronic mild stress (CMS) and determine the alteration in gamma-aminobutyric acid (GABA)-related transmission. Ovx rats were randomly assigned into four groups: (1) estrogen replacement (E2-NoCMS), (2) estrogen replacement and exposure to CMS (E2-CMS), (3) vehicle (VEH-NoCMS), and (4) vehicle and exposure to CMS (VEH-CMS). Following 4-week CMS, VEH groups (VEH-NoCMS and VEH-CMS) showed a similar level of anxiety-like behavior in elevated T-maze, whereas E2-CMS, VEH-NoCMS and VEH-CMS showed anxiety-like behavior in open field. The depressive-like behavior in the force swimming test tended to be affected by estrogen deprivation than CMS. The alteration of the GABAergic system as determined from the GABA level and mRNA expression of GABA-related transmission (i.e., glutamic acid decarboxylase, GABA transporter and GABAA subunits) showed that the GABA level in the amygdala and frontal cortex was affected by CMS. For mRNA expression, the mRNA profile in the amygdala and hippocampus of VEH-NoCMS and E2-CMS was the same but different from those of VEH-NoCMS and E2-CMS. In addition, compared with E2-NoCMS, the mRNA profile in the frontal cortex was similar in VEH-NoCMS, E2-CMS, and VEH-CMS. These findings indicated that the underlying mechanism of the GABAergic system was differently modified, although VEH-NoCMS and VEH-CMS showed anxiety-like behavior. The findings of this study may provide a comprehensive understanding of the modulation of the GABAergic system during estrogen deprivation under CMS, as observed in menopausal women who were daily exposed to stress.
Collapse
Affiliation(s)
- Sushawadee Tongta
- Department of Veterinary Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Suwaporn Daendee
- College of Medicine and Public Health, Ubon Ratchathani University, Ubon Ratchathani 34190, Thailand
| | | |
Collapse
|
20
|
Diviccaro S, FitzGerald JA, Cioffi L, Falvo E, Crispie F, Cotter PD, O’Mahony SM, Giatti S, Caruso D, Melcangi RC. Gut Steroids and Microbiota: Effect of Gonadectomy and Sex. Biomolecules 2022; 12:biom12060767. [PMID: 35740892 PMCID: PMC9220917 DOI: 10.3390/biom12060767] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 01/02/2023] Open
Abstract
Sex steroids, derived mainly from gonads, can shape microbiota composition; however, the impact of gonadectomy and sex on steroid production in the gut (i.e., gut steroids), and its interaction with microbiota composition, needs to be clarified. In this study, steroid environment and gut steroidogenesis were analysed by liquid chromatography tandem mass spectrometry and expression analyses. Gut microbiota composition as branched- and short-chain fatty acids were determined by 16S rRNA gene sequence analysis and gas chromatography flame ionisation detection, respectively. Here, we first demonstrated that levels of pregnenolone (PREG), progesterone (PROG), and isoallopregnanolone (ISOALLO) were higher in the female rat colon, whereas the level of testosterone (T) was higher in males. Sexual dimorphism on gut steroidogenesis is also reported after gonadectomy. Sex, and more significantly, gonadectomy, affects microbiota composition. We noted that a number of taxa and inferred metabolic pathways were associated with gut steroids, such as positive associations between Blautia with T, dihydroprogesterone (DHP), and allopregnanolone (ALLO), whereas negative associations were noted between Roseburia and T, ALLO, PREG, ISOALLO, DHP, and PROG. In conclusion, this study highlights the novel sex-specific association between microbiota and gut steroids with possible relevance for the gut-brain axis.
Collapse
Affiliation(s)
- Silvia Diviccaro
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (E.F.); (S.G.); (D.C.)
| | - Jamie A. FitzGerald
- Teagasc Food Research Centre, Moorepark, P61 C996 Cork, Ireland; (J.A.F.); (F.C.); (P.D.C.)
- APC Microbiome Ireland, University College Cork, College Road, T12 YT20 Cork, Ireland;
| | - Lucia Cioffi
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (E.F.); (S.G.); (D.C.)
| | - Eva Falvo
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (E.F.); (S.G.); (D.C.)
| | - Fiona Crispie
- Teagasc Food Research Centre, Moorepark, P61 C996 Cork, Ireland; (J.A.F.); (F.C.); (P.D.C.)
- APC Microbiome Ireland, University College Cork, College Road, T12 YT20 Cork, Ireland;
| | - Paul D. Cotter
- Teagasc Food Research Centre, Moorepark, P61 C996 Cork, Ireland; (J.A.F.); (F.C.); (P.D.C.)
- APC Microbiome Ireland, University College Cork, College Road, T12 YT20 Cork, Ireland;
| | - Siobhain M. O’Mahony
- APC Microbiome Ireland, University College Cork, College Road, T12 YT20 Cork, Ireland;
| | - Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (E.F.); (S.G.); (D.C.)
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (E.F.); (S.G.); (D.C.)
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (E.F.); (S.G.); (D.C.)
- Correspondence:
| |
Collapse
|
21
|
Diviccaro S, Cioffi L, Falvo E, Giatti S, Melcangi RC. Allopregnanolone: An overview on its synthesis and effects. J Neuroendocrinol 2022; 34:e12996. [PMID: 34189791 PMCID: PMC9285581 DOI: 10.1111/jne.12996] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 12/23/2022]
Abstract
Allopregnanolone, a 3α,5α-progesterone metabolite, acts as a potent allosteric modulator of the γ-aminobutyric acid type A receptor. In the present review, the synthesis of this neuroactive steroid occurring in the nervous system is discussed with respect to physiological and pathological conditions. In addition, its physiological and neuroprotective effects are also reported. Interestingly, the levels of this neuroactive steroid, as well as its effects, are sex-dimorphic, suggesting a possible gender medicine based on this neuroactive steroid for neurological disorders. However, allopregnanolone presents low bioavailability and extensive hepatic metabolism, limiting its use as a drug. Therefore, synthetic analogues or a different therapeutic strategy able to increase allopregnanolone levels have been proposed to overcome any pharmacokinetic issues.
Collapse
Affiliation(s)
- Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanoItaly
| | - Lucia Cioffi
- Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanoItaly
| | - Eva Falvo
- Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanoItaly
| | - Silvia Giatti
- Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanoItaly
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanoItaly
| |
Collapse
|
22
|
Aspesi D, Choleris E. Neuroendocrine underpinning of social recognition in males and females. J Neuroendocrinol 2022; 34:e13070. [PMID: 34927288 DOI: 10.1111/jne.13070] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 11/14/2021] [Accepted: 11/24/2021] [Indexed: 12/28/2022]
Abstract
Social recognition is an essential skill for the expression of appropriate behaviors towards conspecifics in most social species. Several studies point to oxytocin (OT) and arginine vasopressin (AVP) as key mediators of social recognition in males and females. However, sex differences in social cognitive behaviors highlight an important interplay between OT, AVP and the sex steroids. Estrogens facilitate social recognition by regulating OT action in the hypothalamus and that of OT receptor in the medial amygdala. The role of OT in these brain regions appears to be essential for social recognition in both males and females. Conversely, social recognition in male rats and mice is more dependent on AVP release in the lateral septum than in females. The AVP system comprises a series of highly sexually dimorphic brain nuclei, including the bed nucleus of the stria terminalis, the amygdala and the lateral septum. Various studies suggest that testosterone and its metabolites, including estradiol, influence social recognition in males by modulating the activity of the AVP at V1a receptor. Intriguingly, both estrogens and androgens can affect social recognition very rapidly, through non-genomic mechanisms. In addition, the androgen metabolites, namely 3α-diol and 3β-diol, may also have an impact on social behaviors either by interacting with the estrogen receptors or through other mechanisms. Overall, the regulation of OT and AVP by sex steroids fine tunes social recognition and the behaviors that depend upon it (e.g., social bond, hierarchical organization, aggression) in a sex-dependent manner. Elucidating the sex-dependent interaction between sex steroids and neuroendocrine systems is essential for understanding sex differences in the normal and abnormal expression of social behaviors.
Collapse
Affiliation(s)
- Dario Aspesi
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
23
|
Chu X, Snoeren E, Södersten P, Ågmo A. Sexual incentive motivation and male and female copulatory behavior in female rats given androgen from postnatal day 20. Physiol Behav 2021; 237:113460. [PMID: 33991538 DOI: 10.1016/j.physbeh.2021.113460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/03/2021] [Accepted: 05/03/2021] [Indexed: 11/20/2022]
Abstract
Masculinization and feminization of rat sexual behavior has been supposed to occur during a short postnatal period. However, much data have made it evident that these processes may continue until adolescence. In the present study, we evaluated whether androgen treatment of females from postnatal day 20 and onwards could alter sexual motivation and behavior in a male direction. Juveniles were ovariectomized on day 20 and concurrently implanted with Silastic capsules containing either testosterone or dihydrotestosterone. Controls were implanted with an empty capsule. Tests for sexual incentive motivation and male sexual behavior were performed every fifth day when the females were between 50 and 75 days of age. At day 80, a test for female sexual behavior was performed. Females treated with testosterone approached a female sexual incentive far more than a male incentive, showing that sexual motivation had been changed in a male-like direction. Dihydrotestosterone had a similar, albeit smaller, effect. Females implanted with an empty capsule approached both incentives equally. Testosterone produced a high level of mounting behavior, whereas intromission-like behavioral patterns were rare and ejaculation-like behavior was absent. In the test for female sexual behavior, the testosterone-treated animals displayed a relatively high lordosis quotient, far above that displayed in females implanted with dihydrotestosterone or an empty capsule. It is concluded that treatment with an aromatizable androgen during the peripubertal-adolescent period masculinizes sexual motivation and partly sexual behavior. A non-aromatizable androgen weakly masculinize sexual motivation without enhancing male sexual behavior. It appears that simultaneous actions on androgen and estrogen receptors are needed for significant masculinization during the period studied here. Since the testosterone-treated females displayed lordosis, sexual behavior was not defeminized. In sum, these results suggest that sexual differentiation continues well into the peripubertal and adolescent periods.
Collapse
Affiliation(s)
- Xi Chu
- Department of Psychology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Eelke Snoeren
- Department of Psychology, University of Tromsø, 9037 Tromsø, Norway
| | - Per Södersten
- Karolinska Institutet, Novum, S-141 01 Hudddinge, Sweden
| | - Anders Ågmo
- Department of Psychology, University of Tromsø, 9037 Tromsø, Norway.
| |
Collapse
|
24
|
Sze Y, Brunton PJ. Effects of prenatal stress on neuroactive steroid responses to acute stress in adult male and female rats. J Neuroendocrinol 2021; 33:e12916. [PMID: 33270955 PMCID: PMC7900968 DOI: 10.1111/jne.12916] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 12/20/2022]
Abstract
Acute swim stress results in the robust production of several neuroactive steroids, which act as mediators of the stress response. These steroids include glucocorticoids, and positive GABAA receptor modulatory steroids such as allopregnanolone and tetrahydrocorticosterone (THDOC), which potentiate inhibitory GABA signalling, thereby playing a role in the negative control of the hypothalamic-pituitary-adrenal (HPA) axis. Prenatally stressed (PNS) offspring exhibit increased vulnerability to stress-related disorders and frequently display exaggerated HPA axis responses to stressors during adulthood, which may be a result of reduced neuroactive steroid production and consequently inhibitory signalling. Here, we investigated whether exposure of rats to prenatal social stress from gestational day 16-20 altered neuroactive steroid production under non-stress conditions and in response to an acute stressor (swim stress) in adulthood. Using liquid chromatography-mass spectrometry, nine neuroactive steroids were quantified (corticosterone, deoxycorticosterone [DOC], dihydrodeoxycorticosterone, THDOC, progesterone, dihydroprogesterone, allopregnanolone, pregnenolone, testosterone) in plasma and in five brain regions (frontal cortex, hypothalamus, amygdala, hippocampus, brainstem) of male and female control and PNS rats. There was no difference in the neuroactive steroid profile between control and PNS rats under basal conditions. The increase in circulating corticosterone induced by acute swim stress was similar in control and PNS offspring. However, greater stress-induced corticosterone and DOC concentrations were observed in the brainstem of male PNS offspring, whereas DOC concentrations were lower in the hippocampus of PNS females compared to controls, following acute stress. Although PNS rats did not show deficits in allopregnanolone responses to acute stress, there were modest deficits in the production of THDOC in the brainstem, amygdala, and frontal cortex of PNS males and in the frontal cortex of PNS females. The data suggest that neuroactive steroid modulation of GABAergic signalling following stress exposure may be affected in a sex- and region-specific manner in PNS offspring.
Collapse
Affiliation(s)
- Ying Sze
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- The Roslin InstituteUniversity of EdinburghEdinburghUK
| | - Paula J. Brunton
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- The Roslin InstituteUniversity of EdinburghEdinburghUK
- Zhejiang University‐University of Edinburgh Joint InstituteHainingChina
| |
Collapse
|
25
|
Physiopathological Role of Neuroactive Steroids in the Peripheral Nervous System. Int J Mol Sci 2020; 21:ijms21239000. [PMID: 33256238 PMCID: PMC7731236 DOI: 10.3390/ijms21239000] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/21/2020] [Accepted: 11/24/2020] [Indexed: 12/21/2022] Open
Abstract
Peripheral neuropathy (PN) refers to many conditions involving damage to the peripheral nervous system (PNS). Usually, PN causes weakness, numbness and pain and is the result of traumatic injuries, infections, metabolic problems, inherited causes, or exposure to chemicals. Despite the high prevalence of PN, available treatments are still unsatisfactory. Neuroactive steroids (i.e., steroid hormones synthesized by peripheral glands as well as steroids directly synthesized in the nervous system) represent important physiological regulators of PNS functionality. Data obtained so far and here discussed, indeed show that in several experimental models of PN the levels of neuroactive steroids are affected by the pathology and that treatment with these molecules is able to exert protective effects on several PN features, including neuropathic pain. Of note, the observations that neuroactive steroid levels are sexually dimorphic not only in physiological status but also in PN, associated with the finding that PN show sex dimorphic manifestations, may suggest the possibility of a sex specific therapy based on neuroactive steroids.
Collapse
|
26
|
Diviccaro S, Giatti S, Borgo F, Falvo E, Caruso D, Garcia-Segura LM, Melcangi RC. Steroidogenic machinery in the adult rat colon. J Steroid Biochem Mol Biol 2020; 203:105732. [PMID: 32777355 DOI: 10.1016/j.jsbmb.2020.105732] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022]
Abstract
Gastrointestinal function is known to be regulated by steroid molecules produced by the gonads, the adrenal glands and the gut microbiota. However, we have a limited knowledge on the functional significance of local steroid production by gastrointestinal tract tissue. On this basis, we have here evaluated, as a first methodological approach, the expression of steroidogenic molecules and the local levels of key steroids in the male rat colon. Our findings indicate that the colon tissue expresses molecules involved in the early steps of steroidogenesis and in the consecutive synthesis and metabolism of steroid hormones, such as progesterone, testosterone and 17β-estradiol. In addition, the levels of the steroid hormone precursor pregnenolone and the levels of active metabolites of progesterone and testosterone, such as dihydroprogesterone, tetrahydroprogesterone, dihydrotestosterone and 17β-estradiol, were higher in colon than in plasma. Higher levels of the androgen metabolite 3α-diol were detected in the colon in comparison with another non-classical steroidogenic tissue, such as the cerebral cortex. These findings suggest the existence of local steroid synthesis and metabolism in the colon, with the production of active steroid metabolites that may impact on the activity of the enteric nervous system and on the composition of the gut microbiota.
Collapse
Affiliation(s)
- S Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - S Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - F Borgo
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - E Falvo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - D Caruso
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - L M 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
| | - R C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy,.
| |
Collapse
|
27
|
Guennoun R. Progesterone in the Brain: Hormone, Neurosteroid and Neuroprotectant. Int J Mol Sci 2020; 21:ijms21155271. [PMID: 32722286 PMCID: PMC7432434 DOI: 10.3390/ijms21155271] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/29/2020] [Accepted: 07/22/2020] [Indexed: 12/19/2022] Open
Abstract
Progesterone has a broad spectrum of actions in the brain. Among these, the neuroprotective effects are well documented. Progesterone neural effects are mediated by multiple signaling pathways involving binding to specific receptors (intracellular progesterone receptors (PR); membrane-associated progesterone receptor membrane component 1 (PGRMC1); and membrane progesterone receptors (mPRs)) and local bioconversion to 3α,5α-tetrahydroprogesterone (3α,5α-THPROG), which modulates GABAA receptors. This brief review aims to give an overview of the synthesis, metabolism, neuroprotective effects, and mechanism of action of progesterone in the rodent and human brain. First, we succinctly describe the biosynthetic pathways and the expression of enzymes and receptors of progesterone; as well as the changes observed after brain injuries and in neurological diseases. Then, we summarize current data on the differential fluctuations in brain levels of progesterone and its neuroactive metabolites according to sex, age, and neuropathological conditions. The third part is devoted to the neuroprotective effects of progesterone and 3α,5α-THPROG in different experimental models, with a focus on traumatic brain injury and stroke. Finally, we highlight the key role of the classical progesterone receptors (PR) in mediating the neuroprotective effects of progesterone after stroke.
Collapse
Affiliation(s)
- Rachida Guennoun
- U 1195 Inserm and University Paris Saclay, University Paris Sud, 94276 Le kremlin Bicêtre, France
| |
Collapse
|
28
|
Bhatt S, Hillmer AT, Girgenti MJ, Rusowicz A, Kapinos M, Nabulsi N, Huang Y, Matuskey D, Angarita GA, Esterlis I, Davis MT, Southwick SM, Friedman MJ, Duman RS, Carson RE, Krystal JH, Pietrzak RH, Cosgrove KP. PTSD is associated with neuroimmune suppression: evidence from PET imaging and postmortem transcriptomic studies. Nat Commun 2020; 11:2360. [PMID: 32398677 PMCID: PMC7217830 DOI: 10.1038/s41467-020-15930-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 03/31/2020] [Indexed: 12/22/2022] Open
Abstract
Despite well-known peripheral immune activation in posttraumatic stress disorder (PTSD), there are no studies of brain immunologic regulation in individuals with PTSD. [11C]PBR28 Positron Emission Tomography brain imaging of the 18-kDa translocator protein (TSPO), a microglial biomarker, was conducted in 23 individuals with PTSD and 26 healthy individuals-with or without trauma exposure. Prefrontal-limbic TSPO availability in the PTSD group was negatively associated with PTSD symptom severity and was significantly lower than in controls. Higher C-reactive protein levels were also associated with lower prefrontal-limbic TSPO availability and PTSD severity. An independent postmortem study found no differential gene expression in 22 PTSD vs. 22 controls, but showed lower relative expression of TSPO and microglia-associated genes TNFRSF14 and TSPOAP1 in a female PTSD subgroup. These findings suggest that peripheral immune activation in PTSD is associated with deficient brain microglial activation, challenging prevailing hypotheses positing neuroimmune activation as central to stress-related pathophysiology.
Collapse
Affiliation(s)
- Shivani Bhatt
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, 06510, USA
| | - Ansel T Hillmer
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Matthew J Girgenti
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Aleksandra Rusowicz
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Michael Kapinos
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - David Matuskey
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Gustavo A Angarita
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Irina Esterlis
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, 06510, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Margaret T Davis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Steven M Southwick
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | | | - Ronald S Duman
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - John H Krystal
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Robert H Pietrzak
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, 06516, USA
| | - Kelly P Cosgrove
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, 06510, USA.
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA.
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA.
- Yale PET Center, Yale School of Medicine, New Haven, CT, 06519, USA.
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, 06516, USA.
| |
Collapse
|
29
|
Giatti S, Diviccaro S, Falvo E, Garcia-Segura LM, Melcangi RC. Physiopathological role of the enzymatic complex 5α-reductase and 3α/β-hydroxysteroid oxidoreductase in the generation of progesterone and testosterone neuroactive metabolites. Front Neuroendocrinol 2020; 57:100836. [PMID: 32217094 DOI: 10.1016/j.yfrne.2020.100836] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/04/2020] [Accepted: 03/18/2020] [Indexed: 12/22/2022]
Abstract
The enzymatic complex 5α-reductase (5α-R) and 3α/3β-hydroxysteroid oxidoreductase (HSOR) is expressed in the nervous system, where it transforms progesterone (PROG) and testosterone (T) into neuroactive metabolites. These metabolites regulate myelination, brain maturation, neurotransmission, reproductive behavior and the stress response. The expression of 5α-R and 3α-HSOR and the levels of PROG and T reduced metabolites show regional and sex differences in the nervous system and are affected by changing physiological conditions as well as by neurodegenerative and psychiatric disorders. A decrease in their nervous tissue levels may negatively impact the course and outcome of some pathological events. However, in other pathological conditions their increased levels may have a negative impact. Thus, the use of synthetic analogues of these steroids or 5α-R modulation have been proposed as therapeutic approaches for several nervous system pathologies. However, further research is needed to fully understand the consequences of these manipulations, in particular with 5α-R inhibitors.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Eva Falvo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
30
|
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
|
31
|
Sze Y, Brunton PJ. Sex, stress and steroids. Eur J Neurosci 2019; 52:2487-2515. [DOI: 10.1111/ejn.14615] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Ying Sze
- Centre for Discovery Brain Sciences University of Edinburgh Edinburgh UK
| | - Paula J. Brunton
- Centre for Discovery Brain Sciences University of Edinburgh Edinburgh UK
- Zhejiang University‐University of Edinburgh Joint Institute Haining Zhejiang China
| |
Collapse
|
32
|
Neurosteroids and neuropathic pain management: Basic evidence and therapeutic perspectives. Front Neuroendocrinol 2019; 55:100795. [PMID: 31562849 DOI: 10.1016/j.yfrne.2019.100795] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/17/2019] [Accepted: 09/24/2019] [Indexed: 01/18/2023]
Abstract
Complex mechanisms involved in neuropathic pain that represents a major health concern make its management complicated. Because neurosteroids are bioactive steroids endogenously synthesized in the nervous system, including in pain pathways, they appear relevant to develop effective treatments against neuropathic pain. Neurosteroids act in paracrine or autocrine manner through genomic mechanisms and/or via membrane receptors of neurotransmitters that pivotally modulate pain sensation. Basic studies which uncovered a direct link between neuropathic pain symptoms and endogenous neurosteroid production/regulation, paved the way for the investigations of neurosteroid therapeutic potential against pathological pain. Concordantly, antinociceptive properties of synthetic neurosteroids were evidenced in humans and animals. Neurosteroids promote peripheral analgesia mediated by T-type calcium and gamma-aminobutyric acid type A channels, counteract chemotherapy-induced neuropathic pain and ameliorate neuropathic symptoms of injured spinal cord animals by stimulating anti-inflammatory, remyelinating and neuroprotective processes. Together, these data open interesting perspectives for neurosteroid-based strategies to manage/alleviate efficiently neuropathic pain.
Collapse
|