Skip to main content

EDITORIAL article

Front. Endocrinol., 27 July 2022
Sec. Neuroendocrine Science
This article is part of the Research Topic Recent Progress and Perspectives in Neurosteroid Research View all 20 articles

Editorial: Recent Progress and Perspectives in Neurosteroid Research

  • 1Université de Rouen Normandie, Mont-Saint-Aignan, France
  • 2International Cancer Laboratory, Tokyo, Japan
  • 3University of British Columbia, Vancouver, BC, Canada
  • 4Hiroshima University, Hiroshima, Japan

The term "Neurosteroids" designates pregnenolone-derived bioactive compounds that are synthesized or catalyzed by neural cells and act locally on the central or peripheral nervous systems. At the molecular level, the actions of neurosteroids are mainly mediated through membrane receptors i.e. via their cognate receptors and/or via allosteric modulation of other receptors. At the cellular level, neurosteroids act as neurohormones, neuromodulators, neurotransmitters and/or neurotrophic factors. At the organismal level, neurosteroids regulate several physiological processes including arousal, sleep, learning, social and sexual behaviors. Thus, neurosteroids are now considered an important class of intercellular/intracellular signaling molecules in the nervous system, in very much the same as neurotransmitters, neuropeptides and growth factors. Not surprisingly, neurosteroids appear to be implicated in a number of pathophysiological conditions such as pain, neurodegenerative diseases, autism, stress, anxiety, depression, etc. Therefore, drugs targeting neurosteroid biosynthetic enzymes or neurosteroid receptors have strong potential for the development of novel therapeutic approaches.

This Research Topic compiles a series of review and original articles that provide a broad view of the current knowledge on the biosynthesis, functional roles and pathophysiological implications of neurosteroids, and highlights new concepts in this field.

We want to dedicate this Research Topic to the memory of our colleague and dear friend Kazuyoshi Tsutsui, an undisputed leader in neurosteroid research who, very sadly, passed away on September 16, 2021.

Progesterone and allopregnanolone are two neuroactive steroids that act primarily through membrane receptors (1). Progesterone can activate five metabotropic membrane receptors belonging to the progestin and adipoQ receptor (PAQR) family that are distinct from the GPCR family (2). Allopregnanolone and its 3β-methylated synthetic analog ganaxolone act as positive allosteric modulators of GABAA receptors (Pinna). The review by Thomas and Pang summarizes the current knowledge on the neuroprotective actions of allopregnanolone and ganaxolone.

Concurrently, Pinna describes the long journey between the discovery of allopregnanolone in the adrenal cortex (3) and the characterization of its anxiolytic and antidepressant properties (4, 5). The recent validation of allopregnanolone-based treatment of postpartum depression (6) opens new avenues for the development of other neurosteroid-derived drugs in neuropsychiatry.

Apolipoprotein A1 regulatory protein-1 (ARP-1), a member of the steroid receptor superfamily whose ligand is unknown (orphan receptor), regulates transcriptional activity of numerous genes including apolipoprotein-encoding genes (7). Using a yeast one-hybrid screening approach, Honda and Harada identified ARP-1 as a transcription factor that binds to a cis-element, aro-AII, responsible for the brain-specific expression of the mouse aromatase gene.

The recent years have seen substantial progress in the understanding of the bidirectional interactions between the hypothalamo-pituitary-adrenal (HPA) and the hypothalamo-pituitary-gonadal (HPG) axes (8). Hamidovic et al. have conducted a meta-analysis of data spanning over half a century of research on plasma cortisol levels in healthy women during the follicular vs. luteal phases of the menstrual cycle. Their study reveals that circulating cortisol levels are higher in the follicular phase. One possible explanation relies on the changes of GABAA receptor-modulating neurosteroids, including allopregnanolone, during the menstrual cycle (9).

GABAergic anesthetic agents such as isoflurane can exert adverse neuroendocrine effects, notably during the neonatal period (10). Li et al. have investigated the effects of sevoflurane on testosterone (T) and its derivative 17β-estradiol (E2) levels in 5-day old rats. They found that sevoflurane causes an increase in T levels in male rats only and an increase in E2 levels in both male and female rats. These data indicate that the adverse effects of general anesthesia at the beginning of the lifespan might be ascribed, at least in part, to a GABAA receptor-mediated increase of plasma sex steroids.

Biosynthesis of estrogens is catalyzed by the enzyme cytochrome P450 aromatase, also called estrogen synthase, that converts T into E2 (11). A recent study has revealed that administration of aromatase inhibitors may impair cognitive functions (12). In this context, Alia-Klein et al. have taken advantage of the availability of positron emission tomography (PET) aromatase radiotracers to correlate cognitive performance with aromatase levels in the human amygdala and thalamus.

During the perinatal period, neuroestrogens play a pivotal role in the sexual differentiation of the brain (13, 14). The review by Tsukahara and Morishita discusses the role of neuroestrogens of testicular origin on two sexually dimorphic brain regions, the preoptic area (POA) and the bed nucleus of the stria terminalis (BNST). Surprisingly, during the peripubertal period, testicular androgens, without aromatization, also contribute to sexual differentiation of the POA and BNST.

There is now strong evidence that estrogens synthesized within the CNS exert a neuroprotective action (15). Reciprocally, traumatic brain injury (TBI) can affect both estrogen biosynthesis and estrogen inactivation in the central nervous system (CNS) (16). Indeed, TBI can modulate various estrogen metabolizing enzymes including aromatase, steroid sulfatase, estrogen sulfotransferase and 17β-hydroxysteroid dehydrogenases.

Neuroactive steroids, like steroid hormones, can act through intracellular receptors (genomic actions) and/or plasma membrane receptors (non-genomic actions) (17). Neurosteroids are not only synthesized in the CNS but also in the peripheral nervous system (PNS) (18). Colciago et al. provide a comprehensive review of the various aspects of neurosteroid actions in the PNS through intracellular receptors, metabotropic receptors (i.e. G protein-coupled receptors) and ionotropic receptors (mainly GABAA receptors).

Various neurosteroids can negatively or positively modulate GABAA receptor activity and can thus act as proconvulsant or anticonvulsant agents (19). In their systematic review, Miziak et al. summarize the diverse effects of endogenous and exogenous neurosteroids on seizure activity in animal models and epileptic patients.

Astrocytes express both cytochrome P450scc and 3β-hydroxysteroid dehydrogenase, the two enzymes that are required for the biosynthesis of progesterone (20). Although estradiol initiates the luteinizing hormone (LH) surge that triggers ovulation and reproduction, estrogens do not act directly on gonadotropin-releasing hormone (GnRH) neurons (21). Sinchak et al. review the evidence that neuroprogesterone synthesized in hypothalamic astrocytes is involved in the estradiol-induced LH surge.

Fish, which exhibit intense aromatase activity in their CNS (22), represent attractive models to investigate the effect of neuroactive steroids on behavior. Silva et al. reviews the contribution of a weakly electric teleost fish, Gymnotus omarorum, to the understanding of the neuroendocrine mechanisms underlying non-breeding aggressive behavior. The data strongly support the involvement of brain-born estrogens in year-long territorial behavior.

In birds, the biosynthesis of various neurosteroids is higher in the pineal gland than in any other brain region. In particular, 7α-hydroxypregnenolone (23) and allopregnanolone (24) are actively produced in the chicken pineal gland. Haraguchi and Tsutsui review the physiological roles played, respectively, by 7α-hydroxypregnenolone and allopregnanolone in the control of locomotor activity and in Purkinje cell survival during development.

In human as in other vertebrates, sex steroids affect multiple neural and behavioral functions (25). Since the menopause transition is associated with a drop in estrogen levels, He et al. have performed functional MRI scan on premenopausal and perimenopausal women to investigate spontaneous brain activity. The results reveal altered brain functions in brain regions implicated in cognition and working memory in perimenopausal women.

The post-menopausal syndrome includes various neuropsychological disorders including depression, anxiety and dementia (26). In order to investigate the role of estrogens in these disorders, Renczès et al. have compared the effects of surgical (ovariectomy) and pharmacological (aromatase inhibitor) treatment on anxiety-like behavior and memory.

Neuroactive estrogenic and androgenic neurosteroids enhance hippocampal memory tasks (27). In their mini-review, Tozzi et al. recapitulate the evidence supporting the involvement of E2 and T in the induction of long-term potentiation (LTP) and long-term depression (LTD), and on dendritic spine formation in different brain areas. The data indicate that, while estrogens induce LTP and androgens induce LTD, both neurosteroids enhance dendritic spine formation.

Do transient changes in hormonal state during the menstrual cycle affect human behaviors? To answer this question, two types of experimental designs can be set up: within-subject designs where the same women are tested during different phases of their cycle or between-subject designs where two groups of women are tested at different cycle phases. Here, Diekhof et al. have performed a between-subject study to explore the effect of hormonal changes during the late follicular phase and the mid luteal phase on avoidance learning capacity, and they have concurrently conducted a meta-analysis of previously reported within-subject studies. Both approaches concur to demonstrate a decline in avoidance learning during the follicular phase compared to the luteal phase.

In conclusion, the contributions gathered in this Research Topic give an overview on recent advances in our understanding of the physiological roles and potential therapeutic implications of neurosteroids. They also highlight the challenges that remain to be addressed for the next decade. It is our hope that the readers will enjoy reading these articles, and that this Research Topic will become a major set of references for all researchers involved in this rapidly expanding field.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Acknowledgments

We want to thank all the authors of this Research Topic for their excellent contributions, and the dedicated reviewers for their insightful comments that helped maintain the articles at the highest standards. We also gratefully acknowledge the excellent secretarial assistance of Mrs. Catherine Beau and the invaluable support of Emilie Schrepfer, Margaret Georgevits and the whole Frontiers staff.

References

1. Guennoun R, Labombarda F, Gonzalez Deniselle MC, Liere P, De Nicola AF, Schumacher M. Progesterone and Allopregnanolone in the Central Nervous System: Response to Injury and Implication for Neuroprotection. J Steroid Biochem Mol Biol (2015) 146:48–61. doi: 10.1016/j.jsbmb.2014.09.001

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Thomas P. Characteristics of Membrane Progestin Receptor Alpha (mPRalpha) and Progesterone Membrane Receptor Component 1 (PGMRC1) and Their roles in Mediating Rapid Progestin Actions. Front Neuroendocrinol (2008) 29:292–312. doi: 10.1016/j.yfrne.2008.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Beall D, Reichstein T. Isolation of Progesterone and Allopregnanolone From the Adrenal. Nature (1938) 142:479.

Google Scholar

4. Meltzer-Brody S, Kanes SJ. Allopregnanolone in Postpartum Depression: Role in Pathophysiology and Treatment. Neurobiol Stress (2020) 12:100212. doi: 10.1016/j.ynstr.2020.100212

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Walton N, Maguire J. Allopregnanolone-Based Treatments for Postpartum Depression: Why/How Do They Work?. Neurobiol Stress (2019) 11:100198. doi: 10.1016/j.ynstr.2019.100198

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Meltzer-Brody S, Colquhoun H, Riesenberg R, Epperson CN, Deligiannidis KM, Rubinow DR, et al. Brexanolone Injection in Post-Partum Depression: two Multicentre, Double-Blind, Randomised, Placebo-Controlled, Phase 3 Trials. Lancet (2018) 392:1058–70. doi: 10.1016/S0140-6736(18)31551-4

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Ladias JA, Karathanasis SK. Regulation of the Apolipoprotein AI Gene by ARP-1, A Novel Member of the Steroid Receptor Superfamily. Science (1991) 251:561–5. doi: 10.1126/science.1899293

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Acevedo-Rodriguez A, Kauffman AS, Cherrington BD, Borges CS, Roepke TA, Laconi M. Emerging Insights Into Hypothalamic-Pituitary-Gonadal Axis Regulation and Interaction With Stress Signalling. J Neuroendocrinol. (2018) 30:e12590. doi: 10.1111/jne.12590

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Gunn BG, Cunningham L, Mitchell SG, Swinny JD, Lambert JJ, Belelli D. GABAA Receptor-Acting Neurosteroids: a Role in the Development and Regulation of the Stress Response. Front Neuroendocrinol. (2015) 36:28–48. doi: 10.1016/j.yfrne.2014.06.001

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Vutskits L, Xie Z. Lasting Impact of General Anaesthesia on the Brain: Mechanisms and Relevance. Nat Rev Neurosci (2016) 17:705–17. doi: 10.1038/nrn.2016.128

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Simpson ER, Clyne C, Rubin G, Boon WC, Robertson K, Britt K, et al. Aromatase–A Brief Overview. Annu Rev Physiol (2002) 64:93–127. doi: 10.1146/annurev.physiol.64.081601.142703

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Rosenfeld CS, Shay DA, Vieira-Potter VJ. Cognitive Effects of Aromatase and Possible Role in Memory Disorders. Front Endocrinol (Lausanne) (2018) 9:610. doi: 10.3389/fendo.2018.00610

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Amateau SK, McCarthy MM. Sexual differentiation of astrocyte morphology in the Developing Rat Preoptic Area. J Neuroendocrinol. (2002) 14:904–10. doi: 10.1046/j.1365-2826.2002.00858.x

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Sano K, Nakata M, Musatov S, Morishita M, Sakamoto T, Tsukahara S, et al. Pubertal Activation of Estrogen Receptor α in the Medial Amygdala Is Essential for the Full Expression of Male Social Behavior in Mice. Proc Natl Acad Sci USA (2016) 113:7632–7. doi: 10.1073/pnas.1524907113

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Pedersen AL, Nelson LH, Saldanha CJ. Pubertal Activation of Estrogen Receptor α in the Medial Amygdala Is Essential for the Full Expression of Male Social Behavior in Mice. Endocrinology (2016) 157:2041–51. doi: 10.1210/en.2015-1991

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Duncan KA. Estrogen Formation and Inactivation Following TBI: What We Know and Where We Could Go. Front Endocrinol (Lausanne) (2020) 11:345. doi: 10.3389/fendo.2020.00345

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Do Rego JL, Seong JY, Burel D, Leprince J, Vaudry D, Luu-The V, et al. Regulation of Neurosteroid Biosynthesis by Neurotransmitters and Neuropeptides. Front Endocrinol (Lausanne) (2012) 3:4. doi: 10.3389/fendo.2012.00004

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Melcangi RC, Magnaghi V, Galbiati M, Martini L. Formation and Effects of Neuroactive Steroids in the Central and Peripheral Nervous System. Int Rev Neurobiol (2001) 46:145–76. doi: 10.1016/s0074-7742(01)46062-4

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Herd MB, Belelli D, Lambert JJ. Neurosteroid Modulation of Synaptic and Extrasynaptic GABA(A) Receptors. Pharmacol Ther (2007) 116:20–34. doi: 10.1016/j.pharmthera.2007.03.007

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Do Rego JL, Seong JY, Burel D, Leprince J, Luu-The V, Tsutsui K, et al. Neurosteroid Biosynthesis: Enzymatic Pathways and Neuroendocrine Regulation by Neurotransmitters and Neuropeptides. Front Neuroendocrinol. (2009) 30:259–301. doi: 10.1016/j.yfrne.2009.05.006

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Wintermantel TM, Campbell RE, Porteous R, Bock D, Gröne HJ, Todman MG, et al. Definition of Estrogen Receptor Pathway Critical for Estrogen Positive Feedback to Gonadotropin-Releasing Hormone Neurons and Fertility. Neuron (2006) 52:271–80. doi: 10.1016/j.neuron.2006.07.023

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Diotel N, Do Rego JL, Anglade I, Vaillant C, Pellegrini E, Vaudry H, et al. The Brain of Teleost Fish, a Source, and a Target of Sexual Steroids. Front Neurosci (2011) 5:137. doi: 10.3389/fnins.2011.00137

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Hatori M, Hirota T, Iitsuka M, Kurabayashi N, Haraguchi S, Kokame K, et al. Light-Dependent and Circadian Clock-Regulated Activation of Sterol Regulatory Element-Binding Protein, X-Box-Binding Protein 1, and Heat Shock Factor Pathways. Proc Natl Acad Sci USA (2011) 108:4864–9. doi: 10.1073/pnas.1015959108

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Haraguchi S, Hara S, Ubuka T, Mita M, Tsutsui K. Possible Role of Pineal Allopregnanolone in Purkinje Cell Survival. Proc Natl Acad Sci USA (2012) 109:21110–5. doi: 10.1073/pnas.1210804109

PubMed Abstract | CrossRef Full Text | Google Scholar

25. McEwen BS, Milner TA. Understanding the Broad Influence of Sex Hormones and Sex Differences in the Brain. J Neurosci Res (2017) 95:24–39. doi: 10.1002/jnr.23809

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Dotlic J, Radovanovic S, Rancic B, Milosevic B, Nicevic S, Kurtagic I, et al. Mental Health Aspect of Quality of Life in the Menopausal Transition. J Psychosom. Obstet. Gynaecol. (2021) 42:40–9. doi: 10.1080/0167482X.2020.1734789

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Jacome LF, Barateli K, Buitrago D, Lema F, Frankfurt M and Luine VN. Gonadal Hormones Rapidly Enhance Spatial Memory and Increase Hippocampal Spine Density in Male Rats. Endocrinology (2016) 157:1357–62. doi: 10.1210/en.2015-1959

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: neurosteroids, neurohormones, neuromodulators, neurotransmitters, neurotrophic factors

Citation: Vaudry H, Ubuka T, Soma KK and Tsutsui K (2022) Editorial: Recent Progress and Perspectives in Neurosteroid Research. Front. Endocrinol. 13:951990. doi: 10.3389/fendo.2022.951990

Received: 24 May 2022; Accepted: 27 May 2022;
Published: 27 July 2022.

Edited and reviewed by:

Jeff M. P. Holly, University of Bristol, United Kingdom

Copyright © 2022 Vaudry, Ubuka, Soma and Tsutsui. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Hubert Vaudry, hubert.vaudry@univ-rouen.fr

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.