1
|
Nesbitt C, Van Der Walt A, Butzkueven H, Cheung AS, Jokubaitis VG. Exploring the role of sex hormones and gender diversity in multiple sclerosis. Nat Rev Neurol 2025; 21:48-62. [PMID: 39658653 DOI: 10.1038/s41582-024-01042-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2024] [Indexed: 12/12/2024]
Abstract
Sex and sex hormones are thought to influence multiple sclerosis (MS) through effects on inflammation, myelination and neurodegeneration, and exogenous hormones have been explored for their therapeutic potential. However, our understanding of how sex hormones influence MS disease processes and outcomes remains incomplete. Furthermore, our current knowledge is derived primarily from studies that focus exclusively on cisgender populations with exclusion of gender-diverse people. Gender-affirming hormone therapy comprising exogenous sex hormones or sex hormone blocking agents are commonly used by transgender and gender-diverse individuals, and it could influence MS risk and outcomes at various stages of disease. A better understanding of the impact and potential therapeutic effects of both endogenous and exogenous sex hormones in MS is needed to improve care and outcomes for cisgender individuals and, moreover, for gender-diverse populations wherein an evidence base does not exist. In this Perspective, we discuss the effects of endogenous and exogenous sex hormones in MS, including their potential therapeutic benefits, and examine both established sex-based dimorphisms and the potential for gender-diverse dimorphisms. We advocate for future research that includes gender-diverse people to enhance our knowledge of the interplay of sex and sex hormones in MS, leading to the development of more effective and inclusive treatment strategies and improvement of care for all individuals with MS.
Collapse
Affiliation(s)
- Cassie Nesbitt
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia.
- Multiple Sclerosis and Neuroimmunology Clinic, Alfred Health, Melbourne, Victoria, Australia.
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia.
| | - Anneke Van Der Walt
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- Multiple Sclerosis and Neuroimmunology Clinic, Alfred Health, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- Multiple Sclerosis and Neuroimmunology Clinic, Alfred Health, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Ada S Cheung
- Trans Health Research Group, Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
| | - Vilija G Jokubaitis
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia.
- Multiple Sclerosis and Neuroimmunology Clinic, Alfred Health, Melbourne, Victoria, Australia.
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia.
| |
Collapse
|
2
|
Valero-Ochando J, Cantó A, López-Pedrajas R, Almansa I, Miranda M. Role of Gonadal Steroid Hormones in the Eye: Therapeutic Implications. Biomolecules 2024; 14:1262. [PMID: 39456195 PMCID: PMC11506707 DOI: 10.3390/biom14101262] [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: 08/31/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Gonadal steroid hormones are critical regulatory substances involved in various developmental and physiological processes from fetal development through adulthood. These hormones, derived from cholesterol, are synthesized primarily by the gonads, adrenal cortex, and placenta. The synthesis of these hormones involves a series of enzymatic steps starting in the mitochondria and includes enzymes such as cytochrome P450 and aromatase. Beyond their genomic actions, which involve altering gene transcription over hours, gonadal steroids also exhibit rapid, nongenomic effects through receptors located on the cell membrane. Additionally, recent research has highlighted the role of these hormones in the central nervous system (CNS). However, the interactions between gonadal steroid hormones and the retina have received limited attention, though it has been suggested that they may play a protective role in retinal diseases. This review explores the synthesis of gonadal hormones, their mechanisms of action, and their potential implications in various retinal and optic nerve diseases, such as glaucoma, age-related macular degeneration (AMD), diabetic retinopathy (DR), or retinitis pigmentosa (RP), discussing both protective and risk factors associated with hormone levels and their therapeutic potential.
Collapse
Affiliation(s)
| | | | | | | | - María Miranda
- Department of Biomedical Sciences, Faculty of Health Sciences, Institute of Biomedical Sciences, Cardenal Herrera-CEU University, CEU Universities, 46115 Valencia, Spain; (J.V.-O.); (A.C.); (R.L.-P.); (I.A.)
| |
Collapse
|
3
|
Wang Y, Riedstra B, Groothuis T. Effects of maternal androgens and their metabolite etiocholanolone on prenatal development in birds. J Exp Biol 2024; 227:jeb247205. [PMID: 39037123 PMCID: PMC11418167 DOI: 10.1242/jeb.247205] [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: 12/15/2023] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Offspring phenotypes can be affected by maternal testosterone and androstenedione (A4), which are considered a tool of mothers to adjust offspring to a fluctuating environment. Yet testosterone and A4 are very rapidly metabolized by developing avian embryos, suggesting that either the maternal testosterone and A4 have potent organizational effects on the embryos extremely early before being metabolized or it is the metabolites that evoke phenotypic variation in the offspring. One of the metabolites, etiocholanolone, increases substantially during early embryonic development and is a likely candidate for mediating maternal effects as it can promote erythropoiesis. To investigate and compare the effects of testosterone and A4 with the possible effects of etiocholanolone during prenatal embryonic development, we increased their levels in black-headed gull eggs (Larus ridibundus), and used sham-injected eggs as controls. This species usually has 3-egg clutches in which maternal androgen levels increase with the egg-laying sequence. We analysed embryonic heart rate, peri-hatching biometric traits, the ratio of white to red blood cells (W/R ratio) and bursa development. We found that testosterone and A4 treatment increased embryonic heart rate irrespective of egg-laying sequence and decreased bill length and W/R ratio, whereas etiocholanolone did not mimic these effects. Instead, etiocholanolone treatment decreased tarsus length and brain mass. Our finding that etiocholanolone does not mimic the effects induced by testosterone and A4 suggests that the embryonic metabolism of maternal testosterone and A4 can potentially diversify the function of these maternal androgens.
Collapse
Affiliation(s)
- Yuqi Wang
- University of Groningen, Groningen Institute for Evolutionary Life Sciences, 9700 AB Groningen, The Netherlands
| | - Bernd Riedstra
- University of Groningen, Groningen Institute for Evolutionary Life Sciences, 9700 AB Groningen, The Netherlands
| | - Ton Groothuis
- University of Groningen, Groningen Institute for Evolutionary Life Sciences, 9700 AB Groningen, The Netherlands
| |
Collapse
|
4
|
Żulińska S, Strosznajder AK, Strosznajder JB. Current View on PPAR-α and Its Relation to Neurosteroids in Alzheimer's Disease and Other Neuropsychiatric Disorders: Promising Targets in a Therapeutic Strategy. Int J Mol Sci 2024; 25:7106. [PMID: 39000217 PMCID: PMC11241121 DOI: 10.3390/ijms25137106] [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] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) may play an important role in the pathomechanism/pathogenesis of Alzheimer's disease (AD) and several other neurological/neuropsychiatric disorders. AD leads to progressive alterations in the redox state, ion homeostasis, lipids, and protein metabolism. Significant alterations in molecular processes and the functioning of several signaling pathways result in the degeneration and death of synapses and neuronal cells, leading to the most severe dementia. Peroxisome proliferator-activated receptor alpha (PPAR-α) is among the processes affected by AD; it regulates the transcription of genes related to the metabolism of cholesterol, fatty acids, other lipids and neurotransmission, mitochondria biogenesis, and function. PPAR-α is involved in the cholesterol transport to mitochondria, the substrate for neurosteroid biosynthesis. PPAR-α-coding enzymes, such as sulfotransferases, which are responsible for neurosteroid sulfation. The relation between PPAR-α and cholesterol/neurosteroids may have a significant impact on the course and progression of neurodegeneration/neuroprotection processes. Unfortunately, despite many years of intensive studies, the pathogenesis of AD is unknown and therapy for AD and other neurodegenerative diseases is symptomatic, presenting a significant goal and challenge today. This review presents recent achievements in therapeutic approaches for AD, which are targeting PPAR-α and its relation to cholesterol and neurosteroids in AD and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sylwia Żulińska
- Department of Cellular Signaling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| | - Anna K. Strosznajder
- Department of Psychiatry, Medical University of Warsaw, Nowowiejska St. 27, 00-665 Warsaw, Poland;
| | - Joanna B. Strosznajder
- Department of Cellular Signaling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| |
Collapse
|
5
|
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
|
6
|
Yoon S, Kim YK. Endocrinological Treatment Targets for Depressive Disorder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1456:3-25. [PMID: 39261421 DOI: 10.1007/978-981-97-4402-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Depressive disorder exhibits heterogeneity in clinical presentation, progression, and treatment outcomes. While conventional antidepressants based on the monoamine hypothesis benefit many patients, a significant proportion remains unresponsive or fails to fully recover. An individualized integrative treatment approach, considering diverse pathophysiologies, holds promise for these individuals. The endocrine system, governing physiological regulation and organ homeostasis, plays a pivotal role in central nervous system functions. Dysregulations in endocrine system are major cause of depressive disorder due to other medical conditions. Subtle endocrine abnormalities, such as subclinical hypothyroidism, are associated with depression. Conversely, depressive disorder correlates with endocrine-related biomarkers. Fluctuations in sex hormone levels related to female reproduction, elevate depression risk in susceptible subjects. Consequently, extensive research has explored treatment strategies involving the endocrine system. Treatment guidelines recommend tri-iodothyronine augmentation for resistant depression, while allopregnanolone analogs have gained approval for postpartum depression, with ongoing investigations for broader depressive disorders. This book chapter will introduce the relationship between the endocrine system and depressive disorders, presenting clinical findings on neuroendocrinological treatments for depression.
Collapse
Affiliation(s)
- Seoyoung Yoon
- Department of Psychiatry, Daegu Catholic University School of Medicine, Daegu, Republic of Korea
| | - Yong-Ku Kim
- Department of Psychiatry, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
7
|
Han R, Han G, Yan Y, Han L, Li L, Zhang H. Protective effects and mechanisms of the Erzhi formula on glucocorticoid induced primary cortical neuron injury. Front Pharmacol 2023; 14:1038492. [PMID: 36923359 PMCID: PMC10008893 DOI: 10.3389/fphar.2023.1038492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 02/14/2023] [Indexed: 03/03/2023] Open
Abstract
High concentrations of glucocorticoids (GC) can cross the blood-brain barrier into the brain parenchyma, triggering a stress state that can lead to a range of physiological changes. This study investigated whether Erzhi formula has neuroprotective effects against glucocorticoid damage by establishing a dexamethasone-induced primary cortical neuron injury model in vitro. The results showed that Erzhi formula could reduce dexamethasone-induced apoptosis in primary cultured cortical neurons and improve synaptic damage. Further, network pharmacological analysis revealed that Erzhi formula may exert antidepressant effects by multi-component, multi-target, and multi-pathway characteristics, in which Salidroside, Biochanin-A and other ingredients are key components, HSD11B1, NR3C1, and other proteins are key targets, and steroid metabolism may be a key process in its action. Moreover, our study found that the neuroprotective effect of Erzhi formula might be related to the 11β-HSD1-GC/glucocorticoid receptor (GR) signaling pathway. The Erzhi formula could significantly inhibit the activity of 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) in vitro using homogeneous time-resolved fluorescence. In addition to providing evidence for the pharmacological effects of the Erzhi formula, the present study lays down the foundation for subsequent experiments.
Collapse
Affiliation(s)
- Rui Han
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin, China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guoying Han
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin, China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yiqi Yan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin, China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lifeng Han
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin, China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Han Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin, China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
8
|
Kohtz AS, Frye CA. It is all About the Chase: Neurosteroidogenesis in Male Rats is Driven by Control of Mating Pace. Curr Neuropharmacol 2023; 21:1606-1616. [PMID: 36278466 PMCID: PMC10472806 DOI: 10.2174/1570159x21666221019114535] [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: 02/26/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Masculine sexual behaviors are dependent on androstane-derived steroids; however, the modulatory effects of mating, and of mating control, on androstane neurosteroidogenesis remain largely unknown. OBJECTIVE Herein, we investigated the effects of mating control, prior sexual experience, and age on brain region specific neurosteroidogenic responses in male rats. METHODS Effects of acute sexual experience were tested in naïve male rats that either remained sexually- naïve, were exposed to a standard mating chamber, or were either given control of the mating pace in a standard mating chamber (male control) or mated wherein the female stimulus rat controlled the mating pace in a paced-mating chamber (female control). Aged (10-12 months) sexually responsive male rats were similarly euthanized from the homecage or engaged in male controlled or female controlled mating. All rats were euthanized immediately following exposure conditions for radioimmunoassay of steroids in midbrain, hypothalamus, hippocampus and cortex. RESULTS Consummatory sexual behavior in male vs. female-controlled mating paradigms was altered by age and prior sexual experience. Male-controlled mating increased androstane neurosteroid metabolism, such that complementary increases in the testosterone (T) metabolite 5α-androstane-3α-17β- diol (3α-diol) in the midbrain and hypothalamus of male rats corresponded to decreases in the prohormone, T. 3α-diol were increased in the hippocampus in response to the context alone, and to a lesser degree in response to mating. Mating diminished neurosteroidogenesis in the cortex. Neurosteroidogenesis was overall reduced in aged male rats compared to naïve controls, however, these effects were more prominent in sexually non-responsive aged male rats. CONCLUSION Extending previous findings, these results indicate differential production of androstane neurosteroids in a mating exposure, age and brain region dependent manner.
Collapse
Affiliation(s)
- Amy S. Kohtz
- Department of Psychiatry & Human Behavior, Division of Neurobiology & Behavior Research, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, USA
| | - Cheryl A. Frye
- Comprehensive Neuropsychological Services, PLLC, 490 Western Avenue, Albany, NY 12203, USA
| |
Collapse
|
9
|
Pillerová M, Pastorek M, Borbélyová V, Riljak V, Frick KM, Hodosy J, Tóthová L. Sex steroid hormones in depressive disorders as a basis for new potential treatment strategies. Physiol Res 2022; 71:S187-S202. [PMID: 36647907 PMCID: PMC9906660 DOI: 10.33549/physiolres.935001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/17/2022] [Indexed: 01/25/2023] Open
Abstract
The sex steroid hormones (SSHs) such as testosterone, estradiol, progesterone, and their metabolites have important organizational and activational impacts on the brain during critical periods of brain development and in adulthood. A variety of slow and rapid mechanisms mediate both organizational and activational processes via intracellular or membrane receptors for SSHs. Physiological concentrations and distribution of SSHs in the brain result in normal brain development. Nevertheless, dysregulation of hormonal equilibrium may result in several mood disorders, including depressive disorders, later in adolescence or adulthood. Gender differences in cognitive abilities, emotions as well as the 2-3 times higher prevalence of depressive disorders in females, were already described. This implies that SSHs may play a role in the development of depressive disorders. In this review, we discuss preclinical and clinical studies linked to SSHs and development of depressive disorders. Our secondary aim includes a review of up-to-date knowledge about molecular mechanisms in the pathogenesis of depressive disorders. Understanding these molecular mechanisms might lead to significant treatment adjustments for patients with depressive disorders and to an amelioration of clinical outcomes for these patients. Nevertheless, the impact of SSHs on the brain in the context of the development of depressive disorders, progression, and treatment responsiveness is complex in nature, and depends upon several factors in concert such as gender, age, comorbidities, and general health conditions.
Collapse
Affiliation(s)
- M Pillerová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovak Republic.
| | | | | | | | | | | | | |
Collapse
|
10
|
Fernandez N, Petit A, Pianos A, Haddad L, Schumacher M, Liere P, Guennoun R. Aging Is Associated With Lower Neuroactive Steroids and Worsened Outcomes Following Cerebral Ischemia in Male Mice. Endocrinology 2022; 164:6779564. [PMID: 36306407 DOI: 10.1210/endocr/bqac183] [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: 08/08/2022] [Indexed: 01/16/2023]
Abstract
Ischemic stroke is a leading cause of disability and death, and aging is the main nonmodifiable risk factor. Following ischemia, neuroactive steroids have been shown to play a key role in cerebroprotection. Thus, brain steroid concentrations at the time of injury as well as their regulation after stroke are key factors to consider. Here, we investigated the effects of age and cerebral ischemia on steroid levels, behavioral outcomes, and neuronal degeneration in 3- and 18-month-old C57BL/6JRj male mice. Ischemia was induced by middle cerebral artery occlusion for 1 hour followed by reperfusion (MCAO/R) and analyses were performed at 6 hours after MCAO. Extended steroid profiles established by gas chromatography coupled with tandem mass spectrometry revealed that (1) brain and plasma concentrations of the main 5α-reduced metabolites of progesterone, 11-deoxycorticosterone, and corticosterone were lower in old than in young mice; (2) after MCAO/R, brain concentrations of progesterone, 5α-dihydroprogesterone, and corticosterone increased in young mice; and (3) after MCAO/R, brain concentrations of 5α-reduced metabolites of progesterone, 3α5α-tetrahydrodeoxycorticosterone, and 3β5α-tetrahydrodeoxycorticosterone were lower in old than in young mice. After ischemia, old mice showed increased sensori-motor deficits and more degenerating neurons in the striatum than young mice. Altogether, these findings strongly suggest that the decreased capacity of old mice to metabolize steroids toward the 5α-reduction pathway comparatively to young mice may contribute to the worsening of their stroke outcomes.
Collapse
Affiliation(s)
- Neïké Fernandez
- U1195 Inserm and University Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Anthony Petit
- U1195 Inserm and University Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Antoine Pianos
- U1195 Inserm and University Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Léna Haddad
- U1195 Inserm and University Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Michael Schumacher
- U1195 Inserm and University Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Philippe Liere
- U1195 Inserm and University Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Rachida Guennoun
- U1195 Inserm and University Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| |
Collapse
|
11
|
Szczurowska E, Szánti-Pintér E, Randáková A, Jakubík J, Kudova E. Allosteric Modulation of Muscarinic Receptors by Cholesterol, Neurosteroids and Neuroactive Steroids. Int J Mol Sci 2022; 23:13075. [PMID: 36361865 PMCID: PMC9656441 DOI: 10.3390/ijms232113075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/24/2023] Open
Abstract
Muscarinic acetylcholine receptors are membrane receptors involved in many physiological processes. Malfunction of muscarinic signaling is a cause of various internal diseases, as well as psychiatric and neurologic conditions. Cholesterol, neurosteroids, neuroactive steroids, and steroid hormones are molecules of steroid origin that, besides having well-known genomic effects, also modulate membrane proteins including muscarinic acetylcholine receptors. Here, we review current knowledge on the allosteric modulation of muscarinic receptors by these steroids. We give a perspective on the research on the non-genomic effects of steroidal compounds on muscarinic receptors and drug development, with an aim to ultimately exploit such knowledge.
Collapse
Affiliation(s)
- Ewa Szczurowska
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo Namesti 2, Prague 6, 166 10 Prague, Czech Republic
| | - Eszter Szánti-Pintér
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo Namesti 2, Prague 6, 166 10 Prague, Czech Republic
| | - Alena Randáková
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Jan Jakubík
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Eva Kudova
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo Namesti 2, Prague 6, 166 10 Prague, Czech Republic
| |
Collapse
|
12
|
Vitku J, Hill M, Kolatorova L, Kubala Havrdova E, Kancheva R. Steroid Sulfation in Neurodegenerative Diseases. Front Mol Biosci 2022; 9:839887. [PMID: 35281259 PMCID: PMC8904904 DOI: 10.3389/fmolb.2022.839887] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
Steroid sulfation and desulfation participates in the regulation of steroid bioactivity, metabolism and transport. The authors focused on sulfation and desulfation balance in three neurodegenerative diseases: Alzheimer´s disease (AD), Parkinson´s disease (PD), and multiple sclerosis (MS). Circulating steroid conjugates dominate their unconjugated counterparts, but unconjugated steroids outweigh their conjugated counterparts in the brain. Apart from the neurosteroid synthesis in the central nervous system (CNS), most brain steroids cross the blood-brain barrier (BBB) from the periphery and then may be further metabolized. Therefore, steroid levels in the periphery partly reflect the situation in the brain. The CNS steroids subsequently influence the neuronal excitability and have neuroprotective, neuroexcitatory, antidepressant and memory enhancing effects. They also exert anti-inflammatory and immunoprotective actions. Like the unconjugated steroids, the sulfated ones modulate various ligand-gated ion channels. Conjugation by sulfotransferases increases steroid water solubility and facilitates steroid transport. Steroid sulfates, having greater half-lives than their unconjugated counterparts, also serve as a steroid stock pool. Sulfotransferases are ubiquitous enzymes providing massive steroid sulfation in adrenal zona reticularis and zona fasciculata.. Steroid sulfatase hydrolyzing the steroid conjugates is exceedingly expressed in placenta but is ubiquitous in low amounts including brain capillaries of BBB which can rapidly hydrolyze the steroid sulfates coming across the BBB from the periphery. Lower dehydroepiandrosterone sulfate (DHEAS) plasma levels and reduced sulfotransferase activity are considered as risk factors in AD patients. The shifted balance towards unconjugated steroids can participate in the pathophysiology of PD and anti-inflammatory effects of DHEAS may counteract the MS.
Collapse
Affiliation(s)
- Jana Vitku
- Department of Steroids and Proteofactors, Institute of Endocrinology, Prague, Czechia
- *Correspondence: Jana Vitku,
| | - Martin Hill
- Department of Steroids and Proteofactors, Institute of Endocrinology, Prague, Czechia
| | - Lucie Kolatorova
- Department of Steroids and Proteofactors, Institute of Endocrinology, Prague, Czechia
| | - Eva Kubala Havrdova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Radmila Kancheva
- Department of Steroids and Proteofactors, Institute of Endocrinology, Prague, Czechia
| |
Collapse
|
13
|
Sheng JA, Tan SML, Hale TM, Handa RJ. Androgens and Their Role in Regulating Sex Differences in the Hypothalamic/Pituitary/Adrenal Axis Stress Response and Stress-Related Behaviors. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2022; 2:261-274. [PMID: 35024695 PMCID: PMC8744007 DOI: 10.1089/andro.2021.0021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/24/2021] [Indexed: 02/06/2023]
Abstract
Androgens play a pivotal role during development. These gonadal hormones and their receptors exert organizational actions that shape brain morphology in regions controlling the stress regulatory systems in a male-specific manner. Specifically, androgens drive sex differences in the hypothalamic/pituitary/adrenal (HPA) axis and corresponding hypothalamic neuropeptides. While studies have examined the role of estradiol and its receptors in sex differences in the HPA axis and associated behaviors, the role of androgens remains far less studied. Androgens are generally thought to modulate the HPA axis through the activation of androgen receptors (ARs). They can also impact the HPA axis through reduction to estrogenic metabolites that can bind estrogen receptors in the brain and periphery. Such regulation of the HPA axis stress response by androgens can often result in sex-biased risk factors for stress-related disorders, such as anxiety and depression. This review focuses on the biosynthesis pathways and molecular actions of androgens and their nuclear receptors. The impact of androgens on hypothalamic neuropeptide systems (corticotropin-releasing hormone, arginine vasopressin, oxytocin, dopamine, and serotonin) that control the stress response and stress-related disorders is discussed. Finally, this review discusses potential therapeutics involving androgens (androgen replacement therapies, selective AR modulator therapies) and ongoing clinical trials.
Collapse
Affiliation(s)
- Julietta A Sheng
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Sarah M L Tan
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Taben M Hale
- Department of Basic Medical Science, University of Arizona College of Medicine - Phoenix, Arizona, USA
| | - Robert J Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
14
|
Pillerová M, Borbélyová V, Pastorek M, Riljak V, Hodosy J, Frick KM, Tóthová L. Molecular actions of sex hormones in the brain and their potential treatment use in anxiety disorders. Front Psychiatry 2022; 13:972158. [PMID: 36159923 PMCID: PMC9492942 DOI: 10.3389/fpsyt.2022.972158] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Anxiety disorders are one of the most prevalent mood disorders that can lead to impaired quality of life. Current treatment of anxiety disorders has various adverse effects, safety concerns, or restricted efficacy; therefore, novel therapeutic targets need to be studied. Sex steroid hormones (SSHs) play a crucial role in the formation of brain structures, including regions of the limbic system and prefrontal cortex during perinatal development. In the brain, SSHs have activational and organizational effects mediated by either intracellular or transmembrane G-protein coupled receptors. During perinatal developmental periods, the physiological concentrations of SSHs lead to the normal development of the brain; however, the early hormonal dysregulation could result in various anxiety diorders later in life. Sex differences in the prevalence of anxiety disorders suggest that SSHs might be implicated in their development. In this review, we discuss preclinical and clinical studies regarding the role of dysregulated SSHs signaling during early brain development that modifies the risk for anxiety disorders in a sex-specific manner in adulthood. Moreover, our aim is to summarize potential molecular mechanisms by which the SSHs may affect anxiety disorders in preclinical research. Finally, the potential effects of SSHs in the treatment of anxiety disorders are discussed.
Collapse
Affiliation(s)
- Miriam Pillerová
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Veronika Borbélyová
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Michal Pastorek
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Vladimír Riljak
- First Faculty of Medicine, Institute of Physiology, Charles University, Prague, Czechia
| | - Július Hodosy
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - L'ubomíra Tóthová
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
15
|
From Menopause to Neurodegeneration-Molecular Basis and Potential Therapy. Int J Mol Sci 2021; 22:ijms22168654. [PMID: 34445359 PMCID: PMC8395405 DOI: 10.3390/ijms22168654] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
The impacts of menopause on neurodegenerative diseases, especially the changes in steroid hormones, have been well described in cell models, animal models, and humans. However, the therapeutic effects of hormone replacement therapy on postmenopausal women with neurodegenerative diseases remain controversial. The steroid hormones, steroid hormone receptors, and downstream signal pathways in the brain change with aging and contribute to disease progression. Estrogen and progesterone are two steroid hormones which decline in circulation and the brain during menopause. Insulin-like growth factor 1 (IGF-1), which plays an import role in neuroprotection, is rapidly decreased in serum after menopause. Here, we summarize the actions of estrogen, progesterone, and IGF-1 and their signaling pathways in the brain. Since the incidence of Alzheimer’s disease (AD) is higher in women than in men, the associations of steroid hormone changes and AD are emphasized. The signaling pathways and cellular mechanisms for how steroid hormones and IGF-1 provide neuroprotection are also addressed. Finally, the molecular mechanisms of potential estrogen modulation on N-methyl-d-aspartic acid receptors (NMDARs) are also addressed. We provide the viewpoint of why hormone therapy has inconclusive results based on signaling pathways considering their complex response to aging and hormone treatments. Nonetheless, while diagnosable AD may not be treatable by hormone therapy, its preceding stage of mild cognitive impairment may very well be treatable by hormone therapy.
Collapse
|
16
|
Schönfeld P, Reiser G. How the brain fights fatty acids' toxicity. Neurochem Int 2021; 148:105050. [PMID: 33945834 DOI: 10.1016/j.neuint.2021.105050] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 12/24/2022]
Abstract
Neurons spurn hydrogen-rich fatty acids for energizing oxidative ATP synthesis, contrary to other cells. This feature has been mainly attributed to a lower yield of ATP per reduced oxygen, as compared to glucose. Moreover, the use of fatty acids as hydrogen donor is accompanied by severe β-oxidation-associated ROS generation. Neurons are especially susceptible to detrimental activities of ROS due to their poor antioxidative equipment. It is also important to note that free fatty acids (FFA) initiate multiple harmful activities inside the cells, particularly on phosphorylating mitochondria. Several processes enhance FFA-linked lipotoxicity in the cerebral tissue. Thus, an uptake of FFA from the circulation into the brain tissue takes place during an imbalance between energy intake and energy expenditure in the body, a situation similar to that during metabolic syndrome and fat-rich diet. Traumatic or hypoxic brain injuries increase hydrolytic degradation of membrane phospholipids and, thereby elevate the level of FFA in neural cells. Accumulation of FFA in brain tissue is markedly associated with some inherited neurological disorders, such as Refsum disease or X-linked adrenoleukodystrophy (X-ALD). What are strategies protecting neurons against FFA-linked lipotoxicity? Firstly, spurning the β-oxidation pathway in mitochondria of neurons. Secondly, based on a tight metabolic communication between neurons and astrocytes, astrocytes donate metabolites to neurons for synthesis of antioxidants. Further, neuronal autophagy of ROS-emitting mitochondria combined with the transfer of degradation-committed FFA for their disposal in astrocytes, is a potent protective strategy against ROS and harmful activities of FFA. Finally, estrogens and neurosteroids are protective as triggers of ERK and PKB signaling pathways, consequently initiating the expression of various neuronal survival genes via the formation of cAMP response element-binding protein (CREB).
Collapse
Affiliation(s)
- Peter Schönfeld
- Institut für Biochemie und Zellbiologie, Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, D-39120, Magdeburg, Germany
| | - Georg Reiser
- Institut für Inflammation und Neurodegeneration (Neurobiochemie), Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, D-39120, Magdeburg, Germany.
| |
Collapse
|
17
|
Singh R, Bansal R. 16-Substituted steroids alleviate LPS-induced neurodegenerative disorders in rats. Eur J Pharmacol 2021; 895:173876. [PMID: 33460614 DOI: 10.1016/j.ejphar.2021.173876] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/23/2020] [Accepted: 01/11/2021] [Indexed: 10/22/2022]
Abstract
The neuroprotective effects of some 16-substituted steroidal derivatives against the locomotive impairment and cognitive deficits in the lipopolysaccharide (LPS)-induced neuroinflammation model of rats have been investigated. The in vivo and in vitro evaluations include behavioural tests (actophotometer, block tests, Morris water maize and elevated plus maize), estimation of the biochemical parameters such as acetylcholinesterase, lipid peroxide, reactive oxygen, and nitric oxide species and molecular assays for the key proinflammatory mediators like Tumour Necrosis Factor alpha (TNF-α) and Interleukin 1 beta (IL- 1β) after 21 days of the treatment with the steroids. Behavioural and biochemical studies indicated impairment in the locomotor activity and cognitive dysfunction in rats after LPS treatment. In addition, higher levels of TNF-α and IL-1β in the blood serum of the rats were also noticed. However, significant alleviation of LPS-induced movement and memory disorders was observed in LPS-injected rats after treatment with 16-substituted steroidal derivatives 1-11. Furthermore the biochemical and molecular studies revealed suppression of oxidative and nitrosative stress, decreased acetylcholinesterase activity, and reduction of TNF-α and IL-1β levels after treatment with compounds 1-11. Among all the 16-substituted steroidal derivatives, the compounds 8 and 11 were found to be the most active neuroprotective agents and produced effects marginally better than standard drug dexamethasone.
Collapse
Affiliation(s)
- Ranjit Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Ranju Bansal
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| |
Collapse
|
18
|
Gurvich C, Thomas N. Hormones and Cognition. Brain Sci 2021; 11:brainsci11030318. [PMID: 33802267 PMCID: PMC7998363 DOI: 10.3390/brainsci11030318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 11/27/2022] Open
Affiliation(s)
- Caroline Gurvich
- Monash Alfred Psychiatry Research Centre, Central Clinical School, Monash University and The Alfred Hospital, Melbourne, VIC 3004, Australia;
- Correspondence:
| | - Natalie Thomas
- Monash Alfred Psychiatry Research Centre, Central Clinical School, Monash University and The Alfred Hospital, Melbourne, VIC 3004, Australia;
- Department of Biochemistry and Pharmacology, Bio21, University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
19
|
Mouton JC, Duckworth RA. Maternally derived hormones, neurosteroids and the development of behaviour. Proc Biol Sci 2021; 288:20202467. [PMID: 33499795 DOI: 10.1098/rspb.2020.2467] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In a wide range of taxa, there is evidence that mothers adaptively shape the development of offspring behaviour by exposing them to steroids. These maternal effects have major implications for fitness because, by shaping early development, they can permanently alter how offspring interact with their environment. However, theory on parent-offspring conflict and recent physiological studies showing that embryos rapidly metabolize maternal steroids have placed doubt on the adaptive significance of these hormone-mediated maternal effects. Reconciling these disparate perspectives requires a mechanistic understanding of the pathways by which maternal steroids can influence neural development. Here, we highlight recent advances in developmental neurobiology and psychiatric pharmacology to show that maternal steroid metabolites can have direct neuro-modulatory effects potentially shaping the development of neural circuitry underlying ecologically relevant behavioural traits. The recognition that maternal steroids can act through a neurosteroid pathway has critical implications for our understanding of the ecology and evolution of steroid-based maternal effects. Overall, compared to the classic view, a neurosteroid mechanism may reduce the evolutionary lability of hormone-mediated maternal effects owing to increased pleiotropic constraints and frequently influence long-term behavioural phenotypes in offspring.
Collapse
Affiliation(s)
- James C Mouton
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ 85721, USA.,Migratory Bird Center, Smithsonian Conservation Biology Institute, National Zoological Park, MRC 5503, Washington, DC 20013-7012, USA
| | - Renée A Duckworth
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
20
|
Wilson HA, Creighton C, Scharfman H, Choleris E, MacLusky NJ. Endocrine Insights into the Pathophysiology of Autism Spectrum Disorder. Neuroscientist 2020; 27:650-667. [PMID: 32912048 DOI: 10.1177/1073858420952046] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Autism spectrum disorder (ASD) is a class of neurodevelopmental disorders that affects males more frequently than females. Numerous genetic and environmental risk factors have been suggested to contribute to the development of ASD. However, no one factor can adequately explain either the frequency of the disorder or the male bias in its prevalence. Gonadal, thyroid, and glucocorticoid hormones all contribute to normal development of the brain, hence perturbations in either their patterns of secretion or their actions may constitute risk factors for ASD. Environmental factors may contribute to ASD etiology by influencing the development of neuroendocrine and neuroimmune systems during early life. Emerging evidence suggests that the placenta may be particularly important as a mediator of the actions of environmental and endocrine risk factors on the developing brain, with the male being particularly sensitive to these effects. Understanding how various risk factors integrate to influence neural development may facilitate a clearer understanding of the etiology of ASD.
Collapse
Affiliation(s)
- Hayley A Wilson
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada.,Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada
| | - Carolyn Creighton
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Helen Scharfman
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York University Langone Health, New York, NY, USA.,Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Elena Choleris
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada
| | - Neil J MacLusky
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
21
|
Frantsiyants EM, Bandovkina VA, Kaplieva IV, Cheryarina ND, Surikova EI, Neskubina IV, Kotieva IM, Shalashnaya EV, Trepitaki LK. [Influence of malignant growth and chronic neurogenic pain on neurosteroid levels in rat brain]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2020; 66:151-155. [PMID: 32420896 DOI: 10.18097/pbmc20206602151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The aim of the study was to determine the level of sex steroid hormones in white matter of the brain of rats with tumors combined with chronic neurogenic pain (CNP), which was modeled by bilateral sciatic nerve ligation. The study included albino male rats (n=74). In the main group, M1 sarcoma was transplanted subcutaneously (n=11) or into the subclavian vein (n=11) 45 days after CNP modeling. Two comparison groups (n=13 each) included sham operated animals (without CNP) with M1 sarcoma transplanted subcutaneously and intravenously. Control groups included animals with CNP and sham operated animals. Rats were euthanized on day 21 of the carcinogenesis. Levels of total and free testosterone (T), estrone (E1), estradiol (E2), estriol (E3) and progesterone (P4) in the brain white matter were measured using ELISA kits ("Cusabio", China). CNP caused a decrease in the total and free T by 1.5 times (p<0.05), E2 and P4 by 1.9 and 3 times, respectively, E3 by 1.6 times (p<0.05), as well as an increase in E1 by 1.4 times (p<0.05) as compared to the corresponding levels in the brain white matter of rats without CNP. CNP stimulated M1 sarcoma growth in both subcutaneous and intravenous transplantation. Regardless of the tumor site, the dynamics of total T, E2 and E3 in the brain had similar features, but the dynamics of free T, P4 and E1 differed. Thus, changes in the level of neurosteroids in the white matter of rat brain with CNP and tumor growth alone or associated with CNP are a reaction to stress.
Collapse
Affiliation(s)
- E M Frantsiyants
- National Medical Research Centre for Oncology, Rostov-on-Don, Russia
| | - V A Bandovkina
- National Medical Research Centre for Oncology, Rostov-on-Don, Russia
| | - I V Kaplieva
- National Medical Research Centre for Oncology, Rostov-on-Don, Russia
| | - N D Cheryarina
- National Medical Research Centre for Oncology, Rostov-on-Don, Russia
| | - E I Surikova
- National Medical Research Centre for Oncology, Rostov-on-Don, Russia
| | - I V Neskubina
- National Medical Research Centre for Oncology, Rostov-on-Don, Russia
| | - I M Kotieva
- National Medical Research Centre for Oncology, Rostov-on-Don, Russia
| | - E V Shalashnaya
- National Medical Research Centre for Oncology, Rostov-on-Don, Russia
| | - L K Trepitaki
- National Medical Research Centre for Oncology, Rostov-on-Don, Russia
| |
Collapse
|
22
|
McLeod VM, Chiam MDF, Lau CL, Rupasinghe TW, Boon WC, Turner BJ. Dysregulation of Steroid Hormone Receptors in Motor Neurons and Glia Associates with Disease Progression in ALS Mice. Endocrinology 2020; 161:5867502. [PMID: 32621747 DOI: 10.1210/endocr/bqaa113] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/30/2020] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease targeting motor neurons which shows sexual dimorphism in its incidence, age of onset, and progression rate. All steroid hormones, including androgens, estrogens, and progestogens, have been implicated in modulating ALS. Increasing evidence suggests that steroid hormones provide neuroprotective and neurotrophic support to motor neurons, either directly or via surrounding glial cell interactions, by activating their respective nuclear hormone receptors and initiating transcriptional regulatory responses. The SOD1G93A transgenic mouse also shows sex-specific differences in age of onset and progression, and remains the most widely used model in ALS research. To provide a more comprehensive understanding of the influences of steroid hormone signaling in ALS, we systemically characterized sex hormone receptor expression at transcript and protein levels, cellular localization, and the impact of disease course in lumbar spinal cords of male and female SOD1G93A mice. We found that spinal motor neurons highly express nuclear androgen receptor (AR), estrogen receptor (ER)α, ERβ, and progesterone receptor with variations in glial cell expression. AR showed the most robust sex-specific difference in expression and was downregulated in male SOD1G93A mouse spinal cord, in association with depletion in 5α-reductase type 2 isoform, which primarily metabolizes testosterone to 5α-dihydrotestosterone. ERα was highly enriched in reactive astrocytes of SOD1G93A mice and ERβ was strongly upregulated. The 5α-reductase type 1 isoform was upregulated with disease progression and may influence local spinal cord hormone levels. In conclusion, steroid hormone receptor expression is dynamic and cell-type specific in SOD1G93A mice which may provide targets to modulate progression in ALS.
Collapse
Affiliation(s)
- Victoria M McLeod
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Mathew D F Chiam
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Chew L Lau
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Thusitha W Rupasinghe
- Metabolomics Australia, School of BioSciences, University of Melbourne, VIC, Australia
| | - Wah C Boon
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Bradley J Turner
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Perron Institute for Neurological and Translational Science, Queen Elizabeth Medical Centre, Nedlands, WA, Australia
| |
Collapse
|
23
|
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: 80] [Impact Index Per Article: 16.0] [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
|
24
|
Rustichelli C, Bellei E, Bergamini S, Monari E, Baraldi C, Castro FL, Tomasi A, Ferrari A. Serum levels of allopregnanolone, progesterone and testosterone in menstrually-related and postmenopausal migraine: A cross-sectional study. Cephalalgia 2020; 40:1355-1362. [PMID: 32588652 PMCID: PMC7575305 DOI: 10.1177/0333102420937742] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Reduced blood or cerebrospinal fluid levels of allopregnanolone are involved in menstrual cycle-linked CNS disorders, such as catamenial epilepsy. This condition, like menstrually-related migraine, is characterized by severe, treatment-resistant attacks. We explored whether there were differences in allopregnanolone, progesterone and testosterone serum levels between women with menstrually-related migraine (MM, n = 30) or postmenopausal migraine without aura who had suffered from menstrually-related migraine during their fertile age (PM, n = 30) and non-headache control women in fertile age (FAC, n = 30) or post-menopause (PC, n = 30). Methods Participants were women with migraine afferent to a headache centre; controls were female patients’ acquaintances. Serum samples obtained were analyzed by HPLC-ESI-MS/MS. Results In menstrually-related migraine and postmenopausal migraine groups, allopregnanolone levels were lower than in the respective control groups (fertile age and post-menopause) (p < 0.001, one-way analysis of variance followed by Tukey-Kramer post-hoc comparison test) while progesterone and testosterone levels were similar. By grouping together patients with migraine, allopregnanolone levels were inversely correlated with the number of years and days of migraine/3 months (p ≤ 0.005, linear regression analysis). Conclusion Decreased GABAergic inhibition, due to low allopregnanolone serum levels, could contribute to menstrually-related migraine and persistence of migraine after menopause. For the management of these disorders, a rise in the GABAergic transmission by increasing inhibitory neurosteroids might represent a novel strategy.
Collapse
Affiliation(s)
- Cecilia Rustichelli
- Department of Life Sciences, 9306University of Modena and Reggio Emilia, Modena, Italy
| | - Elisa Bellei
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Transplant Surgery, Oncology and Regenerative Medicine Relevance, 9306University of Modena and Reggio Emilia, Modena, Italy
| | - Stefania Bergamini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Transplant Surgery, Oncology and Regenerative Medicine Relevance, 9306University of Modena and Reggio Emilia, Modena, Italy
| | - Emanuela Monari
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Transplant Surgery, Oncology and Regenerative Medicine Relevance, 9306University of Modena and Reggio Emilia, Modena, Italy
| | - Carlo Baraldi
- Department of Biomedical, Metabolic and Neural Sciences, 9306University of Modena and Reggio Emilia, Modena, Italy
| | - Flavia Lo Castro
- School of Pharmacology and Clinical Toxicology, 9306University of Modena and Reggio Emilia, Modena, Italy
| | - Aldo Tomasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Transplant Surgery, Oncology and Regenerative Medicine Relevance, 9306University of Modena and Reggio Emilia, Modena, Italy
| | - Anna Ferrari
- Unit of Medical Toxicology, Headache Centre and Drug Abuse; Department of Biomedical, Metabolic and Neural Sciences, 9306University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
25
|
Inhibitors of cellular stress overcome acute effects of ethanol on hippocampal plasticity and learning. Neurobiol Dis 2020; 141:104875. [PMID: 32334031 DOI: 10.1016/j.nbd.2020.104875] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/03/2020] [Accepted: 04/20/2020] [Indexed: 01/04/2023] Open
Abstract
Ethanol intoxication can produce marked changes in cognitive function including states in which the ability to learn and remember new information is completely disrupted. These defects likely reflect changes in the synaptic plasticity thought to underlie memory formation. We have studied mechanisms contributing to the adverse effects of ethanol on hippocampal long-term potentiation (LTP) and provided evidence that ethanol-mediated LTP inhibition involves a form of metaplasticity resulting from local metabolism of ethanol to acetaldehyde and untimely activation of N-methyl-d-aspartate receptors (NMDARs), both of which are neuronal stressors. In the present studies, we sought to understand the role of cellular stress in LTP defects, and demonstrate that ethanol's effects on LTP in the CA1 hippocampal region are overcome by agents that inhibit cellular stress responses, including ISRIB, a specific inhibitor of integrated stress responses, and GW3965, an agonist that acts at liver X receptors (LXRs) and dampens cellular stress. The agents that alter LTP inhibition also prevent the adverse effects of acute ethanol on one trial inhibitory avoidance learning. Unexpectedly, we found that the LXR agonist but not ISRIB overcomes effects of ethanol on synaptic responses mediated by N-methyl-d-aspartate receptors (NMDARs). These results have implications for understanding the adverse effects of ethanol and possibly for identifying novel paths to treatments that can prevent or overcome ethanol-induced cognitive dysfunction.
Collapse
|
26
|
Sexual hormones regulate the redox status and mitochondrial function in the brain. Pathological implications. Redox Biol 2020; 31:101505. [PMID: 32201220 PMCID: PMC7212485 DOI: 10.1016/j.redox.2020.101505] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 02/11/2020] [Accepted: 03/09/2020] [Indexed: 12/13/2022] Open
Abstract
Compared to other organs, the brain is especially exposed to oxidative stress. In general, brains from young females tend to present lower oxidative damage in comparison to their male counterparts. This has been attributed to higher antioxidant defenses and a better mitochondrial function in females, which has been linked to neuroprotection in this group. However, these differences usually disappear with aging, and the incidence of brain pathologies increases in aged females. Sexual hormones, which suffer a decrease with normal aging, have been proposed as the key factors involved in these gender differences. Here, we provide an overview of redox status and mitochondrial function regulation by sexual hormones and their influence in normal brain aging. Furthermore, we discuss how sexual hormones, as well as phytoestrogens, may play an important role in the development and progression of several brain pathologies, including neurodegenerative diseases such as Alzheimer's and Parkinson's diseases, stroke or brain cancer. Sex hormones are reduced with aging, especially in females, affecting redox balance. Normal aging is associated to a worse redox homeostasis in the brain. Young females show better mitochondrial function and higher antioxidant defenses. Development of brain pathologies is influenced by sex hormones and phytoestrogens.
Collapse
|
27
|
Huilgol D, Venkataramani P, Nandi S, Bhattacharjee S. Transcription Factors That Govern Development and Disease: An Achilles Heel in Cancer. Genes (Basel) 2019; 10:E794. [PMID: 31614829 PMCID: PMC6826716 DOI: 10.3390/genes10100794] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/05/2019] [Accepted: 10/08/2019] [Indexed: 12/22/2022] Open
Abstract
Development requires the careful orchestration of several biological events in order to create any structure and, eventually, to build an entire organism. On the other hand, the fate transformation of terminally differentiated cells is a consequence of erroneous development, and ultimately leads to cancer. In this review, we elaborate how development and cancer share several biological processes, including molecular controls. Transcription factors (TF) are at the helm of both these processes, among many others, and are evolutionarily conserved, ranging from yeast to humans. Here, we discuss four families of TFs that play a pivotal role and have been studied extensively in both embryonic development and cancer-high mobility group box (HMG), GATA, paired box (PAX) and basic helix-loop-helix (bHLH) in the context of their role in development, cancer, and their conservation across several species. Finally, we review TFs as possible therapeutic targets for cancer and reflect on the importance of natural resistance against cancer in certain organisms, yielding knowledge regarding TF function and cancer biology.
Collapse
Affiliation(s)
- Dhananjay Huilgol
- Bungtown Road, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724, USA.
| | | | - Saikat Nandi
- Bungtown Road, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724, USA.
| | - Sonali Bhattacharjee
- Bungtown Road, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724, USA.
| |
Collapse
|
28
|
Yilmaz C, Karali K, Fodelianaki G, Gravanis A, Chavakis T, Charalampopoulos I, Alexaki VI. Neurosteroids as regulators of neuroinflammation. Front Neuroendocrinol 2019; 55:100788. [PMID: 31513776 DOI: 10.1016/j.yfrne.2019.100788] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/12/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023]
Abstract
Neuroinflammation is a physiological protective response in the context of infection and injury. However, neuroinflammation, especially if chronic, may also drive neurodegeneration. Neurodegenerative diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD) and traumatic brain injury (TBI), display inflammatory activation of microglia and astrocytes. Intriguingly, the central nervous system (CNS) is a highly steroidogenic environment synthesizing steroids de novo, as well as metabolizing steroids deriving from the circulation. Neurosteroid synthesis can be substantially affected by neuroinflammation, while, in turn, several steroids, such as 17β-estradiol, dehydroepiandrosterone (DHEA) and allopregnanolone, can regulate neuroinflammatory responses. Here, we review the role of neurosteroids in neuroinflammation in the context of MS, AD, PD and TBI and describe underlying molecular mechanisms. Moreover, we introduce the concept that synthetic neurosteroid analogues could be potentially utilized for the treatment of neurodegenerative diseases in the future.
Collapse
Affiliation(s)
- Canelif Yilmaz
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Kanelina Karali
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Georgia Fodelianaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas, Heraklion, Greece
| | - Vasileia Ismini Alexaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany.
| |
Collapse
|
29
|
Parakala ML, Zhang Y, Modgil A, Chadchankar J, Vien TN, Ackley MA, Doherty JJ, Davies PA, Moss SJ. Metabotropic, but not allosteric, effects of neurosteroids on GABAergic inhibition depend on the phosphorylation of GABA A receptors. J Biol Chem 2019; 294:12220-12230. [PMID: 31239352 PMCID: PMC6690684 DOI: 10.1074/jbc.ra119.008875] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/28/2019] [Indexed: 11/06/2022] Open
Abstract
Neuroactive steroids (NASs) are synthesized within the brain and exert profound effects on behavior. These effects are primarily believed to arise from the activities of NASs as positive allosteric modulators (PAMs) of the GABA-type A receptor (GABAAR). NASs also activate a family of G protein-coupled receptors known as membrane progesterone receptors (mPRs). Here, using surface-biotinylation assays and electrophysiology techniques, we examined mPRs' role in mediating the effects of NAS on the efficacy of GABAergic inhibition. Selective mPR activation enhanced phosphorylation of Ser-408 and Ser-409 (Ser-408/9) within the GABAAR β3 subunit, which depended on the activity of cAMP-dependent protein kinase A (PKA) and protein kinase C (PKC). mPR activation did not directly modify GABAAR activity and had no acute effects on phasic or tonic inhibition. Instead, mPR activation induced a sustained elevation in tonic current, which was blocked by PKA and PKC inhibition. Substitution of Ser-408/9 to alanine residues also prevented the effects of mPR activation on tonic current. Furthermore, this substitution abolished the effects of sustained NAS exposure on tonic inhibition. Interestingly, the allosteric effects of NAS on GABAergic inhibition were independent of Ser-408/9 in the β3 subunit. Additionally, although allosteric effects of NAS on GABAergic inhibition were sensitive to a recently developed "NAS antagonist," the sustained effects of NAS on tonic inhibition were not. We conclude that metabotropic effects of NAS on GABAergic inhibition are mediated by mPR-dependent modulation of GABAAR phosphorylation. We propose that this mechanism may contribute to the varying behavioral effects of NAS.
Collapse
Affiliation(s)
- Manasa L Parakala
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Yihui Zhang
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Amit Modgil
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Jayashree Chadchankar
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Thuy N Vien
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | | | | | - Paul A Davies
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Stephen J Moss
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111; Department of Neuroscience, Physiology, and Pharmacology, University College, London WC1E 6BT, United Kingdom.
| |
Collapse
|
30
|
Li F, Liu L. Comparison of kainate-induced seizures, cognitive impairment and hippocampal damage in male and female mice. Life Sci 2019; 232:116621. [PMID: 31269415 DOI: 10.1016/j.lfs.2019.116621] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/11/2019] [Accepted: 06/29/2019] [Indexed: 12/22/2022]
Abstract
Kainate (KA) mouse model induced by intraperitoneal injection has been widely used for epilepsy and neurodegeneration studies. KA elicits sustained epileptic activity in mouse brain revealed by recurrent behavioral seizures, deteriorative neurodegeneration and various neurological deficits. However, to date, the vast majority of the studies used male mice only, and few studies on the comparison of brain injury between male and female mice in this model were reported. Epidemiological studies indicate that sex may affect the susceptibility to seizure response and neurodegeneration process. Therefore, this study focused on the effect of sex difference on KA-induced recurrent seizures and mortality, locomotor activity and cognitive impairment, and hippocampal neurodegeneration and reactive gliosis in mice. Our results showed that, compared to females, adult male mice exhibited worse performance in mortality rate, severity of epileptic seizures, and cognitive impairment indicated by novel object recognition task. Unexpectedly, post-KA male and female mice underwent similar decline and recovery of locomotor activity. KA-induced neurodegeneration in the whole hippocampus, particularly in CA1 and CA3 subregions, along with the deteriorative reactive gliosis in astrocytes and microglia, was more severe in males than that in females. These data provided the direct in vivo evidence that indicates the key role of sex difference in studies with KA mouse model, and this could be beneficial for optimizing the design of future studies.
Collapse
Affiliation(s)
- Fengling Li
- Department of Pharmacy, Linyi Tumor Hospital, Linyi, Shandong 276001, China
| | - Lei Liu
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
31
|
Intlekofer KA, Clements K, Woods H, Adams H, Suvorov A, Petersen SL. Progesterone receptor membrane component 1 inhibits tumor necrosis factor alpha induction of gene expression in neural cells. PLoS One 2019; 14:e0215389. [PMID: 31026287 PMCID: PMC6485904 DOI: 10.1371/journal.pone.0215389] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/01/2019] [Indexed: 12/31/2022] Open
Abstract
Progesterone membrane receptor component 1 (Pgrmc1) is a cytochrome b5-related protein with wide-ranging functions studied most extensively in non-neural tissues. We previously demonstrated that Pgrmc1 is widely distributed in the brain with highest expression in the limbic system. To determine Pgrmc1 functions in cells of these regions, we compared transcriptomes of control siRNA-treated and Pgrmc1 siRNA-treated N42 hypothalamic cells using whole genome microarrays. Our bioinformatics analyses suggested that Pgrmc1 plays a role in immune functions and likely regulates proinflammatory cytokine signaling. In follow-up studies, we showed that one of these cytokines, TNFα, increased expression of rtp4, ifit3 and gbp4, genes found on microarrays to be among the most highly upregulated by Pgrmc1 depletion. Moreover, either Pgrmc1 depletion or treatment with the Pgrmc1 antagonist, AG-205, increased both basal and TNFα-induced expression of these genes in N42 cells. TNFα had no effect on levels of Rtp4, Ifit3 or Gbp4 mRNAs in mHippoE-18 hippocampal control cells, but Pgrmc1 knock-down dramatically increased basal and TNFα-stimulated expression of these genes. P4 had no effect on gbp4, ifit3 or rtp4 expression or on the ability of Pgrmc1 to inhibit TNFα induction of these genes. However, a majority of the top upstream regulators of Pgrmc1 target genes were related to synthesis or activity of steroids, including P4, that exert neuroprotective effects. In addition, one of the identified Pgrmc1 targets was Nr4a1, an orphan receptor important for the synthesis of most steroidogenic molecules. Our findings indicate that Pgrmc1 may exert neuroprotective effects by suppressing TNFα-induced neuroinflammation and by regulating neurosteroid synthesis.
Collapse
Affiliation(s)
- Karlie A. Intlekofer
- Department of Veterinary and Animal Sciences, Institute of Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Kelsey Clements
- Department of Veterinary and Animal Sciences, Institute of Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Haley Woods
- Department of Veterinary and Animal Sciences, Institute of Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Hillary Adams
- Department of Veterinary and Animal Sciences, Institute of Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Alexander Suvorov
- Department of Environmental Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Sandra L. Petersen
- Department of Veterinary and Animal Sciences, Institute of Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| |
Collapse
|
32
|
Mendell AL, MacLusky NJ. The testosterone metabolite 3α-androstanediol inhibits oxidative stress-induced ERK phosphorylation and neurotoxicity in SH-SY5Y cells through an MKP3/DUSP6-dependent mechanism. Neurosci Lett 2018; 696:60-66. [PMID: 30552945 DOI: 10.1016/j.neulet.2018.12.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 12/04/2018] [Accepted: 12/07/2018] [Indexed: 01/08/2023]
Abstract
Testosterone exerts neuroprotective effects on the brain, but the mechanisms by which these effects are exerted appear to be different in males and females. While in females they involve local conversion to estradiol, in males they may be androgen receptor-dependent, or mediated through metabolism to neurosteroids such as 5α-androstane-3α,17β-diol (3α-diol), which acts through different mechanisms than testosterone itself. Recently, we demonstrated that 3α-diol can protect neurons and neuronal-like cells against oxidative stress-induced neurotoxicity associated with prolonged phosphorylation of the extracellular signal-regulated kinase (ERK). The mechanism(s) responsible for these effects remain unknown. In the present study, we sought to determine whether the ERK-specific phosphatase, mitogen-activated protein kinase phosphatase 3/dual specificity phosphatase 6 (MKP3/DUSP6), is involved in the cytoprotective effects of 3α-diol in SH-SY5Y human female neuroblastoma cells. 3α-diol inhibited ERK phosphorylation and ameliorated cell death induced by the oxidative stressor hydrogen peroxide (H2O2). These protective effects were significantly reduced by pre-treatment with the MKP3/DUSP6 inhibitor BCI. In addition, H2O2 decreased expression of MKP3/DUSP6, and this was prevented by co-treatment with 3α-diol. These findings suggest that the protective effects of 3α-diol are mediated through regulation of ERK phosphorylation in neurotoxic conditions and indicate that these effects may be exerted through modulation of MKP3/DUSP6. Targeting the regulation of MKP3/DUSP6 may be beneficial in reducing toxicity under conditions of oxidative stress.
Collapse
Affiliation(s)
- Ari Loren Mendell
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G 2W1, Canada.
| | - Neil James MacLusky
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|