1
|
Osborne LM, Etyemez S, Pinna G, Alemani R, Standeven LR, Wang XQ, Payne JL. Neuroactive steroid biosynthesis during pregnancy predicts future postpartum depression: a role for the 3α and/or 3β-HSD neurosteroidogenic enzymes? Neuropsychopharmacology 2025; 50:904-912. [PMID: 39885361 PMCID: PMC12032070 DOI: 10.1038/s41386-025-02052-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/21/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025]
Abstract
Postpartum depression (PPD) affects ~10-15% of childbearing individuals, with deleterious consequences for two generations. Recent research has explored the biological mechanisms of PPD, particularly neuroactive steroids (NAS). We sought here to investigate associations between NAS levels and ratios during pregnancy and the subsequent development of depressive symptoms with postpartum onset. NAS levels and psychological scales were measured in individuals with and without mood disorders at up to eight visits across pregnancy and postpartum. Generalized linear mixed-effects regression models were used to assess relationships in euthymic pregnant individuals between each of the NAS biomarkers and ratios and subsequent PPD. Participants with a one-unit increase in the log isoallopregnanolone/pregnanolone ratio at the third trimester (T3) had higher odds (OR = 1.64, 95% CI: 1.13-2.37, FDR adjusted p = 0.038, C-index = 0.82), and those with a one-unit increase in the log pregnanolone/progesterone ratio at T3 had lower odds (OR = 0.64, 95% CI: 0.47-0.88, FDR adjusted p = 0.036, C-index = 0.82) of developing PPD; those with a one-unit increase in the log progesterone level at T3 had higher odds of developing PPD (OR = 4.00, 95% CI: 1.54-10.37, FDR adjusted p = 0.035, C-index = 0.80). We found key differences in the progesterone metabolic pathway at the third trimester, indicating likely decreased activity/expression of the 3α-HSD enzyme and/or increased activity/expression of 3β-HSD.
Collapse
Affiliation(s)
- Lauren M Osborne
- Department of Obstetrics & Gynecology, Weill Cornell Medical College, New York, NY, USA.
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, USA.
| | - Semra Etyemez
- Department of Obstetrics & Gynecology, Weill Cornell Medical College, New York, NY, USA
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, USA
| | - Graziano Pinna
- Department of Psychiatry, The Psychiatric Institute, University of Illinois, Chicago, IL, USA
| | - Rebecca Alemani
- Department of Psychiatry, The Psychiatric Institute, University of Illinois, Chicago, IL, USA
| | - Lindsay R Standeven
- Department of Psychiatry, University of Colorado School of Medicine, Denver, CO, USA
| | - Xin-Qun Wang
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Jennifer L Payne
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
2
|
Demori I, Losacco S, Giordano G, Mucci V, Blanchini F, Burlando B. Fibromyalgia pathogenesis explained by a neuroendocrine multistable model. PLoS One 2024; 19:e0303573. [PMID: 38990866 PMCID: PMC11238986 DOI: 10.1371/journal.pone.0303573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 04/26/2024] [Indexed: 07/13/2024] Open
Abstract
Fibromyalgia (FM) is a central disorder characterized by chronic pain, fatigue, insomnia, depression, and other minor symptoms. Knowledge about pathogenesis is lacking, diagnosis difficult, clinical approach puzzling, and patient management disappointing. We conducted a theoretical study based on literature data and computational analysis, aimed at developing a comprehensive model of FM pathogenesis and addressing suitable therapeutic targets. We started from the evidence that FM must involve a dysregulation of central pain processing, is female prevalent, suggesting a role for the hypothalamus-pituitary-gonadal (HPG) axis, and is stress-related, suggesting a role for the HP-adrenocortical (HPA) axis. Central pathogenesis was supposed to involve a pain processing loop system including the thalamic ventroposterolateral nucleus (VPL), the primary somatosensory cortex (SSC), and the thalamic reticular nucleus (TRN). For decreasing GABAergic and/or increasing glutamatergic transmission, the loop system crosses a bifurcation point, switching from monostable to bistable, and converging on a high-firing-rate steady state supposed to be the pathogenic condition. Thereafter, we showed that GABAergic transmission is positively correlated with gonadal-hormone-derived neurosteroids, notably allopregnanolone, whereas glutamatergic transmission is positively correlated with stress-induced glucocorticoids, notably cortisol. Finally, we built a dynamic model describing a multistable, double-inhibitory loop between HPG and HPA axes. This system has a high-HPA/low-HPG steady state, allegedly reached in females under combined premenstrual/postpartum brain allopregnanolone withdrawal and stress condition, driving the thalamocortical loop to the high-firing-rate steady state, and explaining the connection between endocrine and neural mechanisms in FM pathogenesis. Our model accounts for FM female prevalence and stress correlation, suggesting the use of neurosteroid drugs as a possible solution to currently unsolved problems in the clinical treatment of the disease.
Collapse
Affiliation(s)
- Ilaria Demori
- Department of Pharmacy, DIFAR, University of Genova, Genova, Italy
| | - Serena Losacco
- Department of Pharmacy, DIFAR, University of Genova, Genova, Italy
| | - Giulia Giordano
- Department of Industrial Engineering, University of Trento, Trento, (TN), Italy
- Delft Center for Systems and Control, Delft University of Technology, Delft, The Netherlands
| | - Viviana Mucci
- School of Science, Western Sydney University, Penrith, Australia
| | - Franco Blanchini
- Department of Mathematics, Computer Science and Physics, University of Udine, Udine, Italy
| | - Bruno Burlando
- Department of Pharmacy, DIFAR, University of Genova, Genova, Italy
| |
Collapse
|
3
|
Bengtsson SKS, Sjöstedt J, Malinina E, Das R, Doverskog M, Johansson M, Haage D, Bäckström T. Extra-Synaptic GABA A Receptor Potentiation and Neurosteroid-Induced Learning Deficits Are Inhibited by GR3027, a GABA A Modulating Steroid Antagonist. Biomolecules 2023; 13:1496. [PMID: 37892178 PMCID: PMC10604444 DOI: 10.3390/biom13101496] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Objectives In Vitro: To study the effects of GR3027 (golexanolone) on neurosteroid-induced GABA-mediated current responses under physiological GABAergic conditions with recombinant human α5β3γ2L and α1β2γ2L GABAA receptors expressed in human embryonic kidney cells, using the response patch clamp technique combined with the Dynaflow™ application system. With α5β3γ2L receptors, 0.01-3 μM GR3027, in a concentration-dependent manner, reduced the current response induced by 200 nM THDOC + 0.3 µM GABA, as well as the THDOC-induced direct gated effect. GR3027 (1 μM) alone had no effect on the GABA-mediated current response or current in the absence of GABA. With α1β2γ2L receptors, GR3027 alone had no effect on the GABA-mediated current response or did not affect the receptor by itself. Meanwhile, 1-3 µM GR3027 reduced the current response induced by 200 nM THDOC + 30 µM GABA and 3 µM GR3027 that induced by 200 nM THDOC when GABA was not present. Objectives In Vivo: GR3027 reduces allopregnanolone (AP)-induced decreased learning and anesthesia in male Wistar rats. Rats treated i.v. with AP (2.2 mg/kg) or vehicle were given GR3027 in ratios of 1:0.5 to 1:5 dissolved in 10% 2-hydroxypropyl-beta-cyclodextrin. A dose ratio of AP:GR3027 of at least 1:2.5 antagonized the AP-induced decreased learning in the Morris Water Mase (MWM) and 1:7.5 antagonized the loss of righting reflex (LoR). GR3027 treatment did not change other functions in the rat compared to the vehicle group. Conclusions: GR3027 functions in vitro as an inhibitor of GABAA receptors holding α5β3γ2L and α1β2γ2L, in vivo, in the rat, as a dose-dependent inhibitor toward AP's negative effects on LoR and learning in the MWM.
Collapse
Affiliation(s)
- Sara K. S. Bengtsson
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Umeå University, SE-901 85 Umeå, Sweden
| | - Jessica Sjöstedt
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Umeå University, SE-901 85 Umeå, Sweden
| | - Evgenya Malinina
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Umeå University, SE-901 85 Umeå, Sweden
- Department of Integrative Medical Biology, Umeå University, SE-901 87 Umeå, Sweden
| | - Roshni Das
- Department of Integrative Medical Biology, Umeå University, SE-901 87 Umeå, Sweden
- Umecrine Cognition AB, SE-171 65 Solna, Sweden
| | | | - Maja Johansson
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Umeå University, SE-901 85 Umeå, Sweden
- Umecrine Cognition AB, SE-171 65 Solna, Sweden
| | - David Haage
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Umeå University, SE-901 85 Umeå, Sweden
- Department of Nursing Sciences, Mid Sweden University, AE-851 70 Sundsvall, Sweden
| | - Torbjörn Bäckström
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Umeå University, SE-901 85 Umeå, Sweden
- Umecrine Cognition AB, SE-171 65 Solna, Sweden
| |
Collapse
|
4
|
Bäckström T, Turkmen S, Das R, Doverskog M, Blackburn TP. The GABA system, a new target for medications against cognitive impairment-Associated with neuroactive steroids. J Intern Med 2023; 294:281-294. [PMID: 37518841 DOI: 10.1111/joim.13705] [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] [Indexed: 08/01/2023]
Abstract
The prevalence of cognitive dysfunction, dementia, and neurodegenerative disorders such as Alzheimer's disease (AD) is increasing in parallel with an aging population. Distinct types of chronic stress are thought to be instrumental in the development of cognitive impairment in central nervous system (CNS) disorders where cognitive impairment is a major unmet medical need. Increased GABAergic tone is a mediator of stress effects but is also a result of other factors in CNS disorders. Positive GABA-A receptor modulating stress and sex steroids (steroid-PAMs) such as allopregnanolone (ALLO) and medroxyprogesterone acetate can provoke impaired cognition. As such, ALLO impairs memory and learning in both animals and humans. In transgenic AD animal studies, continuous exposure to ALLO at physiological levels impairs cognition and increases degenerative AD pathology, whereas intermittent ALLO injections enhance cognition, indicating pleiotropic functions of ALLO. We have shown that GABA-A receptor modulating steroid antagonists (GAMSAs) can block the acute negative cognitive impairment of ALLO on memory in animal studies and in patients with cognitive impairment due to hepatic encephalopathy. Here we describe disorders affected by steroid-PAMs and opportunities to treat these adverse effects of steroid-PAMs with novel GAMSAs.
Collapse
Affiliation(s)
| | - Sahruh Turkmen
- Department of Clinical Sciences, University of Umeå, Umeå, Sweden
| | - Roshni Das
- Department of Clinical Sciences, University of Umeå, Umeå, Sweden
- Umecrine Cognition AB, Solna, Sweden
| | | | | |
Collapse
|
5
|
Bäckström T, Bengtsson SKS, Sjöstedt J, Malinina E, Johansson M, Ragagnin G, Ekberg K, Lundgren P. Isoallopregnanolone Inhibits Estrus Cycle-Dependent Aggressive Behavior. Biomolecules 2023; 13:1017. [PMID: 37371597 PMCID: PMC10296718 DOI: 10.3390/biom13061017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/11/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Among female rats, some individuals show estrus cycle-dependent irritability/aggressive behaviors, and these individual rats may be used as a model for premenstrual dysphoric disorder (PMDD). We wanted to investigate if these behaviors are related to the estrus cycle phase containing moderately increased levels of positive GABA-A receptor-modulating steroids (steroid-PAM), especially allopregnanolone (ALLO), and if the adverse behavior can be antagonized. The electrophysiology studies in this paper show that isoallopregnanolone (ISO) is a GABA-A-modulating steroid antagonist (GAMSA), meaning that ISO can antagonize the agonistic effects of positive GABA-A receptor-modulating steroids in both α1β2γ2L and α4β3δ GABA-A receptor subtypes. In this study, we also investigated whether ISO could antagonize the estrus cycle-dependent aggressive behaviors in female Wistar rats using a resident-intruder test. Our results confirmed previous reports of estrus cycle-dependent behaviors in that 42% of the tested rats showed higher levels of irritability/aggression at diestrus compared to those at estrus. Furthermore, we found that, during the treatment with ISO, the aggressive behavior at diestrus was alleviated to a level comparable to that of estrus. We noticed an 89% reduction in the increase in aggressive behavior at diestrus compared to that at estrus. Vehicle treatment in the same animals showed a minimal effect on the diestrus-related aggressive behavior. In conclusion, we showed that ISO can antagonize Steroid-PAM both in α1β2γ2L and α4β3δ GABA-A receptor subtypes and inhibit estrus cycle-dependent aggressive behavior.
Collapse
Affiliation(s)
- Torbjörn Bäckström
- Umeå Neurosteroid Research Center, Department of Clinical Science, Umeå University, SE-901 85 Umea, Sweden
| | - Sara K. S. Bengtsson
- Umeå Neurosteroid Research Center, Department of Clinical Science, Umeå University, SE-901 85 Umea, Sweden
| | - Jessica Sjöstedt
- Umeå Neurosteroid Research Center, Department of Clinical Science, Umeå University, SE-901 85 Umea, Sweden
| | - Evgenya Malinina
- Umeå Neurosteroid Research Center, Department of Clinical Science, Umeå University, SE-901 85 Umea, Sweden
| | - Maja Johansson
- Umeå Neurosteroid Research Center, Department of Clinical Science, Umeå University, SE-901 85 Umea, Sweden
| | - Gianna Ragagnin
- Umeå Neurosteroid Research Center, Department of Clinical Science, Umeå University, SE-901 85 Umea, Sweden
| | - Karin Ekberg
- Asarina Pharma AB, Fogdevreten 2, SE-171 65 Solna, Sweden
| | - Per Lundgren
- Umeå Neurosteroid Research Center, Department of Clinical Science, Umeå University, SE-901 85 Umea, Sweden
| |
Collapse
|
6
|
Sundström-Poromaa I, Comasco E. New Pharmacological Approaches to the Management of Premenstrual Dysphoric Disorder. CNS Drugs 2023; 37:371-379. [PMID: 37171547 DOI: 10.1007/s40263-023-01004-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/13/2023]
Abstract
Premenstrual symptoms are experienced by many female individuals during their fertile age. Premenstrual dysphoric disorder (PMDD), a sex-specific mood disorder, affects about 5% of female individuals during the luteal phase of the menstrual cycle. Treatment with selective serotonin reuptake inhibitors represents a valid solution to manage PMDD for many, but not all, patients. Owing to maladaptive neural reactivity to gonadal hormone fluctuations, that is, the putative mechanism postulated to underlie PMDD, drugs suppressing or stabilizing such variations have been tested. Recently, a clinically significant reduction in the severity of the mental symptoms of PMDD was observed upon treatment with a selective progesterone receptor modulator (SPRM), as demonstrated when comparing ulipristal acetate with placebo in a randomised controlled trial. Stable and low progesterone levels, with maintained low-medium oestradiol levels, define the endocrine profile of this treatment. Importantly, the efficacy of SPRM treatment was accompanied by negligible side effects. These promising results represent a headway to understanding the mechanisms behind PMDD symptomatology and opening up new solutions in the management of PMDD. They also call for studies on the long-term efficacy, safety, and viability of SPRMs in female individuals during their fertile age to further support the development of targeted management of female's mental ill-health in relation to the menstrual cycle. The present overview thus seeks to inform about current and new pharmacological approaches to the management of premenstrual dysphoric disorder.
Collapse
Affiliation(s)
| | - Erika Comasco
- Department of Women's and Children's Health, Science for Life Laboratory, Uppsala University BMC, POB 593, 75124, Uppsala, Sweden.
| |
Collapse
|
7
|
Stiernman L, Dubol M, Comasco E, Sundström-Poromaa I, Boraxbekk CJ, Johansson M, Bixo M. Emotion-induced brain activation across the menstrual cycle in individuals with premenstrual dysphoric disorder and associations to serum levels of progesterone-derived neurosteroids. Transl Psychiatry 2023; 13:124. [PMID: 37055419 PMCID: PMC10101953 DOI: 10.1038/s41398-023-02424-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 04/15/2023] Open
Abstract
Premenstrual dysphoric disorder (PMDD) is a debilitating disorder characterized by severe mood symptoms in the luteal phase of the menstrual cycle. PMDD symptoms are hypothesized to be linked to an altered sensitivity to normal luteal phase levels of allopregnanolone (ALLO), a GABAA-modulating progesterone metabolite. Moreover, the endogenous 3β-epimer of ALLO, isoallopregnanolone (ISO), has been shown to alleviate PMDD symptoms through its selective and dose-dependent antagonism of the ALLO effect. There is preliminary evidence showing altered recruitment of brain regions during emotion processing in PMDD, but whether this is associated to serum levels of ALLO, ISO or their relative concentration is unknown. In the present study, subjects with PMDD and asymptomatic controls underwent functional magnetic resonance imaging (fMRI) in the mid-follicular and the late-luteal phase of the menstrual cycle. Brain responses to emotional stimuli were investigated and related to serum levels of ovarian steroids, the neurosteroids ALLO, ISO, and their ratio ISO/ALLO. Participants with PMDD exhibited greater activity in brain regions which are part of emotion-processing networks during the late-luteal phase of the menstrual cycle. Furthermore, activity in key regions of emotion processing networks - the parahippocampal gyrus and amygdala - was differentially associated to the ratio of ISO/ALLO levels in PMDD subjects and controls. Specifically, a positive relationship between ISO/ALLO levels and brain activity was found in PMDD subjects, while the opposite was observed in controls. In conclusion, individuals with PMDD show altered emotion-induced brain responses in the late-luteal phase of the menstrual cycle which may be related to an abnormal response to physiological levels of GABAA-active neurosteroids.
Collapse
Affiliation(s)
| | - Manon Dubol
- Department of Women's and Children's Health, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Erika Comasco
- Department of Women's and Children's Health, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Carl-Johan Boraxbekk
- Department of Radiation Sciences, Diagnostic Radiology, Umeå University, Umeå, Sweden
- Institute for Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Danish Research Centre for Magnetic Resonance (DRCMR), Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Institute of Sports Medicine Copenhagen (ISMC) and Department of Neurology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Maja Johansson
- Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | - Marie Bixo
- Department of Clinical Sciences, Umeå University, Umeå, Sweden.
| |
Collapse
|
8
|
Solntseva EI, Bukanova JV, Skrebitsky VG, Kudova E. Pregnane neurosteroids exert opposite effects on GABA and glycine-induced chloride current in isolated rat neurons. Hippocampus 2022; 32:552-563. [PMID: 35703084 DOI: 10.1002/hipo.23449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 11/11/2022]
Abstract
The ability of endogenous neurosteroids (NSs) with pregnane skeleton modified at positions C-3 and C-5 to modulate the functional activity of inhibitory glycine receptors (GlyR) and ionotropic ɣ-aminobutyric acid receptors (GABAA R) was estimated. The glycine and GABA-induced chloride current (IGly and IGABA ) were measured in isolated pyramidal neurons of the rat hippocampus and in isolated rat cerebellar Purkinje cells, respectively. Our experiments demonstrated that pregnane NSs affected IGABA and IGly in a different manner. At low concentrations (up to 5 μM), tested pregnane NSs increased or did not change the peak amplitude of the IGABA , but reduced the IGly by decreasing the peak amplitude and/or accelerating desensitization. Namely, allopregnanolone (ALLO), epipregnanolone (EPI), pregnanolone (PA), pregnanolone sulfate (PAS) and 5β-dihydroprogesterone (5β-DHP) enhanced the IGABA in Purkinje cells. Dose-response curves plotted in the concentration range from 1 nM to 100 μM were smooth for EPI and 5β-DHP, but bell-shaped for ALLO, PA and PAS. The peak amplitude of the IGly was reduced by PA, PAS, and 5α- and 5β-DHP. In contrast, ALLO, ISO and EPI did not modulate it. Dose-response curves for the inhibition of the IGly peak amplitude were smooth for all active compounds. All NSs accelerated desensitization of the IGly . The dose-response relationship for this effect was smooth for ALLO, PA, PAS and 5β-DHP, but it was U-shaped for EPI, 5α-DHP and ISO. These results, together with our previous results on NSs with androstane skeleton, offer comprehensive overview for understanding the mechanisms of effects of NSs on IGly and IGABA .
Collapse
Affiliation(s)
- Elena I Solntseva
- Functional Synaptology Laboratory, Brain Research Department, Research Center of Neurology, Moscow, Russia
| | - Julia V Bukanova
- Functional Synaptology Laboratory, Brain Research Department, Research Center of Neurology, Moscow, Russia
| | - Vladimir G Skrebitsky
- Functional Synaptology Laboratory, Brain Research Department, Research Center of Neurology, Moscow, Russia
| | - Eva Kudova
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
9
|
Feng YH, Lim SW, Lin HY, Wang SA, Hsu SP, Kao TJ, Ko CY, Hsu TI. Allopregnanolone suppresses glioblastoma survival through decreasing DPYSL3 and S100A11 expression. J Steroid Biochem Mol Biol 2022; 219:106067. [PMID: 35114375 DOI: 10.1016/j.jsbmb.2022.106067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 01/15/2022] [Accepted: 01/28/2022] [Indexed: 12/14/2022]
Abstract
Allopregnanolone (allo) is a physiological regulator of neuronal activity that treats multiple neurological disorders. Allo penetrates the blood-brain barrier with very high efficiency, implying that allo can treat CNS-related diseases, including glioblastoma (GBM), which always recurs after standard therapy. Hence, this study aimed to determine whether allo has a therapeutic effect on GBM. We found that allo enhanced temozolomide (TMZ)-suppressed cell survival and proliferation of TMZ-resistant cells. In particular, allo enhanced TMZ-inhibited cell migration and TMZ-induced apoptosis. Additionally, allo strongly induced DNA damage characterized by γH2Ax. Furthermore, quantitative proteomic analysis, iTRAQ, showed that allo significantly decreased the levels of DPYSL3, S100A11, and S100A4, reflecting the poor prognosis of patients with GBM confirmed by differential gene expression and survival analysis. Moreover, single-cell RNA-Seq revealed that S100A11, expressed in malignant cells, oligodendrocytes, and macrophages, was significantly associated with immune cell infiltration. Furthermore, overexpression of DPYSL3 or S100A11 prevented allo-induced cell death. In conclusion, allo suppresses GBM cell survival by decreasing DPYSL3/S100A11 expression and inducing DNA damage.
Collapse
Affiliation(s)
| | - Sher-Wei Lim
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan; Department of Neurosurgery, Chi-Mei Medical Center, Tainan 722, Taiwan; Department of Nursing, Min-Hwei College of Health Care Management, Tainan 736, Taiwan
| | - Hong-Yi Lin
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan
| | - Shao-An Wang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Sung-Po Hsu
- Department of Physiology, School of Medicine, Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Tzu-Jen Kao
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan
| | - Chiung-Yuan Ko
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan.
| | - Tsung-I Hsu
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei 110, Taiwan.
| |
Collapse
|
10
|
Bortolato M, Coffey BJ, Gabbay V, Scheggi S. Allopregnanolone: The missing link to explain the effects of stress on tic exacerbation? J Neuroendocrinol 2022; 34:e13022. [PMID: 34423500 PMCID: PMC8800948 DOI: 10.1111/jne.13022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/19/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022]
Abstract
The neurosteroid allopregnanolone (3α-hydroxy-5α-pregnan-20-one; AP) elicits pleiotropic effects in the central nervous system, ranging from neuroprotective and anti-inflammatory functions to the regulation of mood and emotional responses. Several lines of research show that the brain rapidly produces AP in response to acute stress to reduce the allostatic load and enhance coping. These effects not only are likely mediated by GABAA receptor activation but also result from the contributions of other mechanisms, such as the stimulation of membrane progesterone receptors. In keeping with this evidence, AP has been shown to exert rapid, potent antidepressant properties and has been recently approved for the therapy of moderate-to-severe postpartum depression. In addition to depression, emerging evidence points to the potential of AP as a therapy for other neuropsychiatric disorders, including anxiety, seizures, post-traumatic stress disorder and cognitive problems. Although this evidence has spurred interest in further therapeutic applications of AP, some investigations suggest that this neurosteroid may also be associated with adverse events in specific disorders. For example, our group has recently documented that AP increases tic-like manifestations in several animal models of tic disorders; furthermore, our results indicate that inhibiting AP synthesis and signalling reduces the exacerbation of tic severity associated with acute stress. Although the specific mechanisms of these effects remain partially elusive, our findings point to the possibility that the GABAergic activation by AP may also lead to disinhibitory effects, which could interfere with the ability of patients to suppress their tics. Future studies will be necessary to verify whether these mechanisms may apply to other externalising manifestations, such as impulse-control problems and manic symptoms.
Collapse
Affiliation(s)
- Marco Bortolato
- Department of Pharmacology and ToxicologyCollege of PharmacyUniversity of UtahSalt Lake CityUTUSA
- Research Consortium on NeuroEndocrine Causes of Tics (ReConNECT)
| | - Barbara J. Coffey
- Research Consortium on NeuroEndocrine Causes of Tics (ReConNECT)
- Department of Psychiatry and Behavioral ScienceMiller School of MedicineUniversity of MiamiMiamiFLUSA
| | - Vilma Gabbay
- Research Consortium on NeuroEndocrine Causes of Tics (ReConNECT)
- Department of Psychiatry and Behavioral SciencesAlbert Einstein College of MedicineBronxNYUSA
| | - Simona Scheggi
- Department of Molecular and Developmental MedicineSchool of MedicineUniversity of SienaSienaItaly
| |
Collapse
|
11
|
Bäckström T, Das R, Bixo M. Positive GABA A receptor modulating steroids and their antagonists: Implications for clinical treatments. J Neuroendocrinol 2022; 34:e13013. [PMID: 34337790 DOI: 10.1111/jne.13013] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 01/02/2023]
Abstract
GABA is the main inhibitory neurotransmitter in the brain and GABAergic transmission has been shown to be of importance for regulation of mood, memory and food intake. The progesterone metabolite allopregnanolone (Allo) is a positive GABAA receptor modulating steroid with potent effects. In humans, disorders such as premenstrual dysphoric disorder (PMDD), hepatic encephalopathy and polycystic ovarian syndrome are associated with elevated Allo levels and increased negative mood, disturbed memory and increased food intake in some individuals. This is surprising because Allo shares many properties with benzodiazepines and is mainly considered to be anxiolytic and anti-depressant. However, it is well established that, in certain individuals, GABAA receptor activating compounds could have paradoxical effects and thus be anxiogenic in low physiological plasma concentrations but anxiolytic at high levels. We have demonstrated that isoallopregnanolone (Isoallo), the 3β-OH sibling of Allo, functions as a GABAA receptor modulating steroid antagonist (GAMSA) but without any effects of its own on GABAA receptors. The antagonistic effect is noted in most GABAA subtypes investigated in vitro to date. In vivo, Isoallo can inhibit Allo-induced anaesthesia in rats, as well as sedation or saccadic eye velocity in humans. Isoallo treatment has been studied in women with PMDD. In a first phase II study, Isoallo (Sepranolone; Asarina Pharma) injections significantly ameliorated negative mood in women with PMDD compared with placebo. Several GAMSAs for oral administration have also been developed. The GAMSA, UC1011, can inhibit Allo induced memory disturbances in rats and an oral GAMSA, GR3027, has been shown to restore learning and motor coordination in rats with hepatic encephalopathy. In humans, vigilance, cognition and pathological electroencephalogram were improved in patients with hepatic encephalopathy on treatment with GR3027. In conclusion GAMSAs are a new possible treatment for disorders and symptoms caused by hyperactivity in the GABAA system.
Collapse
Affiliation(s)
- Torbjörn Bäckström
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, Umea, Sweden
| | - Roshni Das
- Department of Integrative Medical Biology, Umeå University, Umea, Sweden
| | - Marie Bixo
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, Umea, Sweden
| |
Collapse
|
12
|
Epipregnanolone as a Positive Modulator of GABA A Receptor in Rat Cerebellar and Hippocampus Neurons. Biomolecules 2021; 11:biom11060791. [PMID: 34074021 PMCID: PMC8225200 DOI: 10.3390/biom11060791] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 11/24/2022] Open
Abstract
Epipregnanolone (3β-hydroxy-5β-pregnan-20-one, Epi) is an endogenous steroid with important physiological effects and high affinity for GABAA receptors. The effect of Epi on GABA-induced chloride current (IGABA) in native neurons has hardly been studied. In this work, we studied the influence of Epi on the IGABA in the Purkinje cells of rat cerebellum and pyramidal neurons of rat hippocampus with the patch clamp technique. We showed that Epi is a positive modulator of the IGABA with EC50 of 5.7 µM in Purkinje cells and 9.3 µM in hippocampal neurons. Epi-induced potentiation of the IGABA was more potent at low vs. high GABA concentrations. Isopregnanolone (3β-hydroxy-5α-pregnan-20-one, Iso) counteracted Epi, reducing its potentiating effect by 2–2.3 times. Flumazenil, a nonsteroidal GABAA receptor antagonist, does not affect the Epi-induced potentiation. Comparison of the potentiating effects of Epi and allopregnanolone (3α-hydroxy-5α-pregnan-20-one, ALLO) showed that ALLO is, at least, a four times more potent positive modulator than Epi. The combined application of ALLO and Epi showed that the effects of these two steroids are not additive. We conclude that Epi has a dual effect on the IGABA increasing the current in the control solution and decreasing the stimulatory effect of ALLO.
Collapse
|
13
|
Bengtsson S, Bäckström T, Brinton R, Irwin R, Johansson M, Sjöstedt J, Wang M. GABA-A receptor modulating steroids in acute and chronic stress; relevance for cognition and dementia? Neurobiol Stress 2020; 12:100206. [PMID: 31921942 PMCID: PMC6948369 DOI: 10.1016/j.ynstr.2019.100206] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/13/2019] [Accepted: 12/18/2019] [Indexed: 01/10/2023] Open
Abstract
Cognitive dysfunction, dementia and Alzheimer's disease (AD) are increasing as the population worldwide ages. Therapeutics for these conditions is an unmet need. This review focuses on the role of the positive GABA-A receptor modulating steroid allopregnanolone (APα), it's role in underlying mechanisms for impaired cognition and of AD, and to determine options for therapy of AD. On one hand, APα given intermittently promotes neurogenesis, decreases AD-related pathology and improves cognition. On the other, continuous exposure of APα impairs cognition and deteriorates AD pathology. The disparity between these two outcomes led our groups to analyze the mechanisms underlying the difference. We conclude that the effects of APα depend on administration pattern and that chronic slightly increased APα exposure is harmful to cognitive function and worsens AD pathology whereas single administrations with longer intervals improve cognition and decrease AD pathology. These collaborative assessments provide insights for the therapeutic development of APα and APα antagonists for AD and provide a model for cross laboratory collaborations aimed at generating translatable data for human clinical trials.
Collapse
Affiliation(s)
- S.K.S. Bengtsson
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| | - T. Bäckström
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| | - R. Brinton
- Center for Innovation in Brain Science, Professor Departments of Pharmacology and Neurology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - R.W. Irwin
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089, USA
| | - M. Johansson
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| | - J. Sjöstedt
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| | - M.D. Wang
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| |
Collapse
|
14
|
Cadeddu R, Bäckström T, Floris G, Nordkild P, Segerdahl M, Bortolato M. Isoallopregnanolone reduces tic-like behaviours in the D1CT-7 mouse model of Tourette syndrome. J Neuroendocrinol 2020; 32:e12754. [PMID: 31175669 DOI: 10.1111/jne.12754] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/16/2019] [Accepted: 06/04/2019] [Indexed: 12/29/2022]
Abstract
Tourette syndrome (TS) is a neuropsychiatric disorder characterised by multiple, persistent tics. These semi-voluntary motor and phonic manifestations are typically aggravated by exposure to acute stress, yet the mechanisms underlying this exacerbation remain unclear. Using a well-characterised animal model of TS, the D1CT-7 mouse, we recently showed that acute stress increases tic-like responses and causes sensorimotor gating deficits, as measured by the prepulse inhibition of the startle. We showed that these effects are promoted by the brain synthesis of the neurosteroid allopregnanolone (AP). In line with this idea, inhibition of AP synthesis by finasteride was found to suppress the tic-exacerbating effects of stress; conversely, AP administration resulted in a marked enhancement of the number of tic-like motor bursts. Given that the primary mechanism of AP is based on the positive allosteric modulation of GABAA receptors, in the present study, we hypothesised that the enhancement in tic-like behaviours induced by either stress or AP may be countered by isoallopregnanolone (isoAP), the natural 3β-epimer of AP that acts as an antagonist to the AP-binding site within GABAA receptors. In agreement with our hypothesis, isoAP (5-10 mg kg-1 , s.c.) dose-dependently reduced the number of tic-like behaviours induced by stress in D1CT-7 mice. These effects were comparable to those elicited by both the benchmark TS therapy haloperidol (0.3 mg kg-1 , i.p.), as well as finasteride (25 mg kg-1 , i.p.). IsoAP also countered the prepulse inhibition deficits secondary to stress in D1CT-7 mice. Finally, isoAP opposed the enhancement of tic-like behaviours induced by AP (15 mg kg-1 , i.p.). Given that isoAP is well-tolerated and has an optimal safety profile, these data suggest that this steroid may have therapeutic properties in TS.
Collapse
Affiliation(s)
- Roberto Cadeddu
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Torbjörn Bäckström
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, Umeå, Sweden
- Asarina Pharma, Copenhagen, Denmark
| | - Gabriele Floris
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | | | | | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
15
|
Progestogen profiling in plasma during the estrous cycle in cattle using an LC-MS based approach. Theriogenology 2019; 142:376-383. [PMID: 31708192 DOI: 10.1016/j.theriogenology.2019.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 10/02/2019] [Accepted: 10/08/2019] [Indexed: 11/23/2022]
Abstract
In many mammalian species, corpus luteum derived progesterone (P4) is the main functional gestagen during the estrous cycle and pregnancy. P4 can be metabolized into various metabolites, of which some are biologically active. While some metabolites target the classical nuclear progesterone receptor (PR), neurosteroids bind the receptors of type A γ-aminobutyric acid (GABAA-r) in the brain. According to the position of reduction within the molecule, metabolites of P4 can be characterized into C20-reduced progestogens (20α-dihydroprogesterone (20α-DHP) and 20β-dihydroprogesterone (20β-DHP)), C3-reduced progestogens (3α-dihydroprogesterone (3α-DHP) and 3β-dihydroprogesterone (3β-DHP)), 5α-reduced progestogens (5α-dihydroprogesterone (5α-DHP), allopregnanolone and isopregnanolone) and 5β-reduced progestogens (5β-dihydroprogesterone (5β-DHP), pregnanolone and epipregnanolone). We questioned whether the reduced progestogens are present in bovine plasma during the estrous cycle and whether their profiles differed from the profile of the common precursor P4 around the time of luteolysis. The analytes were monitored in plasma samples using liquid chromatography mass spectrometry (LC-MS). While progestogens lagged behind the drop of P4 at luteolysis, they followed the profile of P4 during the estrous cycle. The abundance of P4 was predominant followed by allopregnanolone, pregnanolone, epipregnanolone and 20β-DHP. Further studies will need to focus particularly on the period around luteolysis.
Collapse
|
16
|
Bixo M, Johansson M, Timby E, Michalski L, Bäckström T. Effects of GABA active steroids in the female brain with a focus on the premenstrual dysphoric disorder. J Neuroendocrinol 2018; 30. [PMID: 29072794 DOI: 10.1111/jne.12553] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/15/2017] [Accepted: 10/21/2017] [Indexed: 12/28/2022]
Abstract
Premenstrual dysphoric disorder (PMDD) afflicts 3%-5% of women of childbearing age, and is characterised by recurrent negative mood symptoms (eg, irritability, depression, anxiety and emotional lability) during the luteal phase of the menstrual cycle. The aetiology of PMDD is unknown, although a temporal association with circulating ovarian steroids, in particular progesterone and its metabolite allopregnanolone, has been established during the luteal phase. Allopregnanolone is a positive modulator of the GABAA receptor: it is sedative in high concentrations but may precipitate paradoxical adverse effects on mood at levels corresponding to luteal phase concentrations in susceptible women. Saccadic eye velocity (SEV) is a measure of GABAA receptor sensitivity; in experimental studies of healthy women, i.v. allopregnanolone decreases SEV. Women with PMDD display an altered sensitivity to an i.v. injection of allopregnanolone compared to healthy controls in this model. In functional magnetic resonance imaging (fMRI) studies, women with PMDD react differently to emotional stimuli in contrast to controls. A consistent finding in PMDD patients is increased amygdala reactivity during the luteal phase. Post-mortem studies in humans have revealed that allopregnanolone concentrations vary across different brain regions, although mean levels in the brain also reflect variations in peripheral serum concentrations. The amygdala processes emotions such as anxiety and aggression. This is interesting because allopregnanolone is detected at high concentrations within the region into which marked increases in blood flow are measured with fMRI following progesterone/allopregnanolone administration. Allopregnanolone effects are antagonised by its isomer isoallopregnanolone (UC1010), which significantly reduces negative mood symptoms in women with PMDD when administered s.c. in the premenstrual phase. This was shown in a randomised, placebo-controlled clinical trial in which the primary outcome was change in symptom scoring on the Daily Rating of Severity of Problems (DRSP): the treatment reduced negative mood scores (P < .005), as well as total DRSP scores (P < .01), compared to placebo in women with PMDD. In conclusion, the underlying studies of this review provide evidence that allopregnanolone is the provoking factor behind the negative mood symptoms in PMDD and that isoallopregnanolone could ameliorate the symptoms as a result of its ability to antagonise the allopregnanolone effect on the GABAA receptor.
Collapse
Affiliation(s)
- M Bixo
- Department of Clinical Science, Obstetrics and Gynecology, Umeå University, Umeå, Sweden
| | - M Johansson
- Department of Clinical Science, Obstetrics and Gynecology, Umeå University, Umeå, Sweden
| | - E Timby
- Department of Clinical Science, Obstetrics and Gynecology, Umeå University, Umeå, Sweden
| | - L Michalski
- Department of Clinical Science, Obstetrics and Gynecology, Umeå University, Umeå, Sweden
| | - T Bäckström
- Department of Clinical Science, Obstetrics and Gynecology, Umeå University, Umeå, Sweden
| |
Collapse
|
17
|
Bixo M, Ekberg K, Poromaa IS, Hirschberg AL, Jonasson AF, Andréen L, Timby E, Wulff M, Ehrenborg A, Bäckström T. Treatment of premenstrual dysphoric disorder with the GABA A receptor modulating steroid antagonist Sepranolone (UC1010)-A randomized controlled trial. Psychoneuroendocrinology 2017; 80:46-55. [PMID: 28319848 DOI: 10.1016/j.psyneuen.2017.02.031] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/07/2017] [Accepted: 02/27/2017] [Indexed: 01/04/2023]
Abstract
CONTEXT Allopregnanolone is a metabolite from progesterone and a positive modulator of the GABAA receptor. This endogenous steroid may induce negative mood in sensitive women when present in serum levels comparable to the premenstrual phase. Its endogenous isomer, isoallopregnanolone, has been shown to antagonize allopregnanolone effects in experimental animal and human models. OBJECTIVE The objective was to test whether inhibition of allopregnanolone by treatment with the GABAA modulating steroid antagonist (GAMSA) Sepranolone (UC1010) during the premenstrual phase could reduce symptoms of the premenstrual dysphoric disorder (PMDD). The pharmacokinetic parameters of UC1010 when given as a subcutaneous injection were measured in healthy women prior to the study in women with PMDD. DESIGN This was an explorative randomized, double-blind, placebo-controlled study. SETTING Swedish multicentre study with 10 centers. PARTICIPANTS Participants were 26 healthy women in a pharmacokinetic phase I study part, and 126 women with PMDD in a phase II study part. Diagnosis followed the criteria for PMDD in DSM-5 using Daily Record of Severity of Problems (DRSP) and Endicott's algorithm. INTERVENTION Subjects were randomized to treatment with UC1010 (10 or 16mg) subcutaneously every second day during the luteal phase or placebo during one menstrual cycle. OUTCOME MEASURES The primary outcome measure was the sum of all 21 items in DRSP (Total DRSP score). Secondary outcomes were Negative mood score i.e. the ratings of the 4 key symptoms in PMDD (anger/irritability, depression, anxiety and lability) and impairment (impact on daily life). RESULTS 26 healthy women completed the pharmacokinetic phase I study and the dosing in the following trial was adjusted according to the results. 106 of the 126 women completed the phase II study. Within this group, a significant treatment effect with UC1010 compared to placebo was obtained for the Total DRSP score (p=0.041) and borderline significance (p=0.051) for the sum of Negative mood score. Nineteen participants however showed symptoms during the follicular phase that might be signs of an underlying other conditions, and 27 participants had not received the medication as intended during the symptomatic phase. Hence, to secure that the significant result described above was not due to chance, a post hoc sub-group analysis was performed, including only women with pure PMDD who completed the trial as intended (n=60). In this group UC1010 reduced Total DRSP scores by 75% compared with 47% following placebo; the effect size 0.7 (p=0.006), and for sum of Negative mood score (p=0.003) and impairment (p=0.010) with the effect size 0.6. No severe adverse events were reported during the treatment and safety parameters (vital signs and blood chemistry) remained normal during the study. CONCLUSIONS This explorative study indicates promising results for UC1010 as a potential treatment for PMDD. The effect size was comparable to that of SSRIs and drospirenone containing oral contraceptives. UC1010 was well tolerated and deemed safe.
Collapse
Affiliation(s)
- Marie Bixo
- Department of Clinical Science, Umeå University, SE-901 85 Umeå, Sweden.
| | - Karin Ekberg
- Asarina Pharma AB, Fogdevreten 2, SE-171 65 Solna, Sweden
| | | | - Angelica Lindén Hirschberg
- Department of Women's and Children's Health, Karolinska Institutet and Department of Obstetrics and Gynecology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Aino Fianu Jonasson
- Kvinnoforskningsenheten, Karolinska University Hospital, SE-141 46 Huddinge, Sweden
| | - Lotta Andréen
- Department of Obstetrics and Gynecology, Sundsvall Hospital, SE-851 86 Sundsvall, Sweden
| | - Erika Timby
- Department of Clinical Science, Umeå University, SE-901 85 Umeå, Sweden
| | - Marianne Wulff
- Slottsstadens Läkarhus Malmö, Fågelbacksgatan 11, SE-217 44 Malmö, Sweden
| | - Agneta Ehrenborg
- Specialistläkarna Kungsbacka (Qvinnolivet), Södra Torggatan 18, SE-434 30 Kungsbacka, Sweden
| | | |
Collapse
|
18
|
Johansson M, Strömberg J, Ragagnin G, Doverskog M, Bäckström T. GABAA receptor modulating steroid antagonists (GAMSA) are functional in vivo. J Steroid Biochem Mol Biol 2016; 160:98-105. [PMID: 26523675 DOI: 10.1016/j.jsbmb.2015.10.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 10/16/2015] [Accepted: 10/25/2015] [Indexed: 12/18/2022]
Abstract
GABAA receptor modulating steroid antagonists (GAMSA) selectively inhibit neurosteroid-mediated enhancement of GABA-evoked currents at the GABAA receptor. 3α-hydroxy-neurosteroids, notably allopregnanolone and tetrahydrodeoxycorticosterone (THDOC), potentiate GABAA receptor-mediated currents. On the contrary, various 3β-hydroxy-steroids antagonize this positive neurosteroid-mediated modulation. Importantly, GAMSAs are specific antagonists of the positive neurosteroid-modulation of the receptor and do not inhibit GABA-evoked currents. Allopregnanolone and THDOC have both negative and positive actions. Allopregnanolone can impair encoding/consolidation and retrieval of memories. Chronic administration of a physiological allopregnanolone concentration reduces cognition in mice models of Alzheimer's disease. In humans an allopregnanolone challenge impairs episodic memory and in hepatic encephalopathy cognitive deficits are accompanied by increased brain ammonia and allopregnanolone. Hippocampal slices react in vitro to ammonia by allopregnanolone synthesis in CA1 neurons, which blocks long-term potentiation (LTP). Thus, allopregnanolone may impair learning and memory by interfering with hippocampal LTP. Contrary, pharmacological treatment with allopregnanolone can promote neurogenesis and positively influence learning and memory of trace eye-blink conditioning in mice. In rat the GAMSA UC1011 inhibits an allopregnanolone-induced learning impairment and the GAMSA GR3027 restores learning and motor coordination in rats with hepatic encephalopathy. In addition, the GAMSA isoallopregnanolone antagonizes allopregnanolone-induced anesthesia in rats, and in humans it antagonizes allopregnanolone-induced sedation and reductions in saccadic eye velocity. 17PA is also an effective GAMSA in vivo, as it antagonizes allopregnanolone-induced anesthesia and spinal analgesia in rats. In vitro the allopregnanolone/THDOC-increased GABA-mediated GABAA receptor activity is antagonized by isoallopregnanolone, UC1011, GR3027 and 17PA, while the effect of GABA itself is not affected.
Collapse
Affiliation(s)
- Maja Johansson
- Umeå Neurosteroid research center, Obstetrics and Gynecology, Clinical Sciences at Umeå University, Building 6M, 4th floor at NUS, SE-901 85 Umeå, Sweden; Umecrine Cognition AB, Sweden.
| | - Jessica Strömberg
- Umeå Neurosteroid research center, Obstetrics and Gynecology, Clinical Sciences at Umeå University, Building 6M, 4th floor at NUS, SE-901 85 Umeå, Sweden
| | - Gianna Ragagnin
- Umeå Neurosteroid research center, Obstetrics and Gynecology, Clinical Sciences at Umeå University, Building 6M, 4th floor at NUS, SE-901 85 Umeå, Sweden
| | | | - Torbjörn Bäckström
- Umeå Neurosteroid research center, Obstetrics and Gynecology, Clinical Sciences at Umeå University, Building 6M, 4th floor at NUS, SE-901 85 Umeå, Sweden
| |
Collapse
|
19
|
Abstract
In certain women, increased negative mood relates to the progesterone metabolite, allopregnanolone (allo), during the luteal phase of ovulatory menstrual cycles, the premenstrual dysphoric disorder (PMDD). In anovulatory cycles, no symptom or sex steroid increase occurs but symptoms return during progesterone/allo treatment. Allo is a potent GABAA receptor-modulating steroid and as such is expected to be calming and anxiolytic. A relation to negative mood is unexpected. However, this paradoxical effect can be induced by all GABAA receptor modulators in low concentrations whereas higher concentrations are calming. The severity of the mood symptoms relate to allo in an inverted U-shaped curve at endogenous luteal-phase serum concentrations. Allo's effects on the GABAA receptor can be antagonized by isoallopregnanolone (ISO), an antagonist to allo. ISO has also been used in a preliminary clinical trial on PMDD ameliorating symptoms with good effect in PMDD patients.
Collapse
Affiliation(s)
- Torbjörn Bäckström
- Department of Clinical sciences, Obstetrics and Gynecology, Umeå neurosteroid research center, Umeå University, Building 6M 4th floor, Umeå, Sweden.
| | - Marie Bixo
- Department of Clinical sciences, Obstetrics and Gynecology, Umeå neurosteroid research center, Umeå University, Building 6M 4th floor, Umeå, Sweden.
| | - Jessica Strömberg
- Department of Clinical sciences, Obstetrics and Gynecology, Umeå neurosteroid research center, Umeå University, Building 6M 4th floor, Umeå, Sweden.
| |
Collapse
|
20
|
Johansson M, Agusti A, Llansola M, Montoliu C, Strömberg J, Malinina E, Ragagnin G, Doverskog M, Bäckström T, Felipo V. GR3027 antagonizes GABAA receptor-potentiating neurosteroids and restores spatial learning and motor coordination in rats with chronic hyperammonemia and hepatic encephalopathy. Am J Physiol Gastrointest Liver Physiol 2015; 309:G400-G409. [PMID: 26138462 PMCID: PMC4556948 DOI: 10.1152/ajpgi.00073.2015] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/29/2015] [Indexed: 01/31/2023]
Abstract
Hepatic encephalopathy (HE) is one of the primary complications of liver cirrhosis. Current treatments for HE, mainly directed to reduction of ammonia levels, are not effective enough because they cannot completely eliminate hyperammonemia and inflammation, which induce the neurological alterations. Studies in animal models show that overactivation of GABAA receptors is involved in cognitive and motor impairment in HE and that reducing this activation restores these functions. We have developed a new compound, GR3027, that selectively antagonizes the enhanced activation of GABAA receptors by neurosteroids such as allopregnanolone and 3α,21-dihydroxy-5α-pregnan-20-one (THDOC). This work aimed to assess whether GR3027 improves motor incoordination, spatial learning, and circadian rhythms of activity in rats with HE. GR3027 was administered subcutaneously to two main models of HE: rats with chronic hyperammonemia due to ammonia feeding and rats with portacaval shunts (PCS). Motor coordination was assessed in beam walking and spatial learning and memory in the Morris water maze and the radial maze. Circadian rhythms of ambulatory and vertical activity were also assessed. In both hyperammonemic and PCS rats, GR3027 restores motor coordination, spatial memory in the Morris water maze, and spatial learning in the radial maze. GR3027 also partially restores circadian rhythms of ambulatory and vertical activity in PCS rats. GR3027 is a novel approach to treatment of HE that would normalize neurological functions altered because of enhanced GABAergic tone, affording more complete normalization of cognitive and motor function than current treatments for HE.
Collapse
Affiliation(s)
- Maja Johansson
- Umecrine Cognition AB, Solna, Sweden; Umeå Neurosteroid Research Center, Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | | | | | | | - Jessica Strömberg
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | - Evgenya Malinina
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | - Gianna Ragagnin
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | | | - Torbjörn Bäckström
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | | |
Collapse
|
21
|
Bengtsson SKS, Nyberg S, Hedström H, Zingmark E, Jonsson B, Bäckström T, Bixo M. Isoallopregnanolone antagonize allopregnanolone-induced effects on saccadic eye velocity and self-reported sedation in humans. Psychoneuroendocrinology 2015; 52:22-31. [PMID: 25459890 DOI: 10.1016/j.psyneuen.2014.10.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/30/2014] [Accepted: 10/31/2014] [Indexed: 01/09/2023]
Abstract
Allopregnanolone (AP) is an endogenous neurosteroid. It modulates the effect of γ-amino-butyric acid (GABA) on the GABA type A (GABAA) receptor, which leads to increased receptor activity. Since the GABA-system is mainly inhibitory, increased AP activity leads to modulation of neuronal activity. In vitro studies of GABAA receptor activity and in vivo animal studies of sedation have shown that AP-induced effects can be inhibited by another endogenous steroid, namely isoallopregnanolone (ISO). In this study we investigated if ISO can antagonize AP-induced effects in healthy female volunteers, via measurements of saccadic eye velocity (SEV) and self-rated sedation. With a single-blind cross-over design, 12 women were studied on three separate occasions; given AP alone or AP in combination with one of two ISO doses. Congruent with previous reports, AP administration decreased SEV and induced sedation and these effects were diminished by simultaneous ISO administration. Also, the ISO effect modulation was seemingly stronger for SEV than for sedation. These effects were observed already at an ISO dose exposure that was approximately half of that of AP. In conclusion, ISO antagonized AP-induced decrease in SEV and self-reported sedation, probably in a non-competitive manner.
Collapse
Affiliation(s)
- Sara K S Bengtsson
- Umeå Neurosteroid Research Center, Department of Clinical Science, Umeå University, Umeå, Sweden
| | - Sigrid Nyberg
- Umeå Neurosteroid Research Center, Department of Clinical Science, Umeå University, Umeå, Sweden
| | - Helena Hedström
- Umeå Neurosteroid Research Center, Department of Clinical Science, Umeå University, Umeå, Sweden
| | - Elisabeth Zingmark
- Umeå Neurosteroid Research Center, Department of Clinical Science, Umeå University, Umeå, Sweden
| | - Björn Jonsson
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Torbjörn Bäckström
- Umeå Neurosteroid Research Center, Department of Clinical Science, Umeå University, Umeå, Sweden.
| | - Marie Bixo
- Umeå Neurosteroid Research Center, Department of Clinical Science, Umeå University, Umeå, Sweden
| |
Collapse
|
22
|
Maldonado-Devincci AM, Beattie MC, Morrow DH, McKinley RE, Cook JB, O’Buckley TK, Morrow AL. Reduction of circulating and selective limbic brain levels of (3α,5α)-3-hydroxy-pregnan-20-one (3α,5α-THP) following forced swim stress in C57BL/6J mice. Psychopharmacology (Berl) 2014; 231:3281-92. [PMID: 24744202 PMCID: PMC4335654 DOI: 10.1007/s00213-014-3552-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/16/2014] [Indexed: 01/12/2023]
Abstract
RATIONALE Stress activates the hypothalamic-pituitary-adrenal (HPA) axis, and GABAergic neuroactive steroids contribute to homeostatic regulation of this circuitry. Acute forced swim stress (FSS) increases plasma, cortical, and hypothalamic (3α,5α)-3-hydroxy-pregnan-20-one (3α,5α-THP) levels in rats. However, there have not been systemic investigations of acute stress on changes in plasma and brain levels of 3α,5α-THP in mouse models. OBJECTIVES The present experiments aimed to assess circulating and local brain levels of 3α,5α-THP following acute FSS in C57BL/6J mice. METHODS Mice were exposed to FSS (10 min), and 50 min later, blood and brains were collected. Circulating pregnenolone and 3α,5α-THP levels were assessed in serum. Free-floating brain sections (40 μm, four to five sections/region) were immunostained and analyzed in cortical and limbic brain structures. RESULTS FSS decreased circulating 3α,5α-THP (-41.6 ± 10.4 %) and reduced 3α,5α-THP immunolabeling in the paraventricular nucleus of the hypothalamus (-15.2 ± 5.7 %), lateral amygdala (LA, -31.1 ± 13.4 %), and nucleus accumbens (NAcc) shell (-31.9 ± 14.6). Within the LA, vesicular glutamate transporter 1 (VGLUT1) and vesicular GABA transporter were localized in 3α,5α-THP-positively stained cells, while in the NAcc shell, only VGLUT1 was localized in 3α,5α-THP-positively stained cells, suggesting that both glutamatergic and GABAergic cells within the LA are 3α,5α-THP-positive, while in the NAcc shell, 3α,5α-THP only localizes to glutamatergic cells. CONCLUSIONS The decrease in circulating and brain levels of 3α,5α-THP may be due to alterations in the biosynthesis/metabolism or changes in the regulation of the HPA axis following FSS. Changes in GABAergic neuroactive steroids in response to stress likely mediate functional adaptations in neuronal activity. This may provide a potential targeted therapeutic avenue to address maladaptive stress responsivity.
Collapse
Affiliation(s)
- Antoniette M. Maldonado-Devincci
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Building, CB 7178, Chapel Hill, NC 27599, USA,Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew C. Beattie
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Building, CB 7178, Chapel Hill, NC 27599, USA
| | - Danielle H. Morrow
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Building, CB 7178, Chapel Hill, NC 27599, USA
| | - Raechel E. McKinley
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Building, CB 7178, Chapel Hill, NC 27599, USA
| | - Jason B. Cook
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Building, CB 7178, Chapel Hill, NC 27599, USA,Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Todd K. O’Buckley
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Building, CB 7178, Chapel Hill, NC 27599, USA
| | - A. Leslie Morrow
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Building, CB 7178, Chapel Hill, NC 27599, USA,Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA,Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA,Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
23
|
di Michele F, Luchetti S, Bernardi G, Romeo E, Longone P. Neurosteroid and neurotransmitter alterations in Parkinson's disease. Front Neuroendocrinol 2013; 34:132-42. [PMID: 23563222 DOI: 10.1016/j.yfrne.2013.03.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 02/17/2013] [Accepted: 03/25/2013] [Indexed: 01/13/2023]
Abstract
Parkinson's disease (PD) is associated with a massive loss of dopaminergic cells in the substantia nigra leading to dopamine hypofunction and alteration of the basal ganglia circuitry. These neurons, are under the control, among others, of the excitatory glutamatergic and inhibitory γ-aminobutyric acid (GABA) systems. An imbalance between these systems may contribute to excitotoxicity and dopaminergic cell death. Neurosteroids, a group of steroid hormones synthesized in the brain, modulate the function of several neurotransmitter systems. The substantia nigra of the human brain expresses high concentrations of allopregnanolone (3α, 5αtetrahydroprogesterone), a neurosteroid that positively modulates the action of GABA at GABAA receptors and of 5α-dihydroprogesterone, a neurosteroid acting at the genomic level. This article reviews the roles of NS acting as neuroprotectants and as GABAA receptor agonists in the physiology and pathophysiology of the basal ganglia, their impact on dopaminergic cell activity and survival, and potential therapeutic application in PD.
Collapse
|
24
|
Estrous cycle variations in GABA(A) receptor phosphorylation enable rapid modulation by anabolic androgenic steroids in the medial preoptic area. Neuroscience 2012; 226:397-410. [PMID: 22989919 DOI: 10.1016/j.neuroscience.2012.09.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 08/31/2012] [Accepted: 09/06/2012] [Indexed: 12/13/2022]
Abstract
Anabolic androgenic steroids (AAS), synthetic testosterone derivatives that are used for ergogenic purposes, alter neurotransmission and behaviors mediated by GABA(A) receptors. Some of these effects may reflect direct and rapid action of these synthetic steroids at the receptor. The ability of other natural allosteric steroid modulators to alter GABA(A) receptor-mediated currents is dependent upon the phosphorylation state of the receptor complex. Here we show that phosphorylation of the GABA(A) receptor complex immunoprecipitated by β(2)/β(3) subunit-specific antibodies from the medial preoptic area (mPOA) of the mouse varies across the estrous cycle; with levels being significantly lower in estrus. Acute exposure to the AAS, 17α-methyltestosterone (17α-MeT), had no effect on the amplitude or kinetics of inhibitory postsynaptic currents in the mPOA of estrous mice when phosphorylation was low, but increased the amplitude of these currents from mice in diestrus, when it was high. Inclusion of the protein kinase C (PKC) inhibitor, calphostin, in the recording pipette eliminated the ability of 17α-MeT to enhance currents from diestrous animals, suggesting that PKC-receptor phosphorylation is critical for the allosteric modulation elicited by AAS during this phase. In addition, a single injection of 17α-MeT was found to impair an mPOA-mediated behavior (nest building) in diestrus, but not in estrus. PKC is known to target specific serine residues in the β(3) subunit of the GABA(A) receptor. Although phosphorylation of these β(3) serine residues showed a similar profile across the cycle, as did phosphoserine in mPOA lysates immunoprecipitated with β2/β3 antibody (lower in estrus than in diestrus or proestrus), the differences were not significant. These data suggest that the phosphorylation state of the receptor complex regulates both the ability of AAS to modulate receptor function in the mPOA and the expression of a simple mPOA-dependent behavior through a PKC-dependent mechanism that involves the β(3) subunit and other sites within the GABA(A) receptor complex.
Collapse
|
25
|
Timby E, Hedström H, Bäckström T, Sundström-Poromaa I, Nyberg S, Bixo M. Allopregnanolone, a GABAA receptor agonist, decreases gonadotropin levels in women. A preliminary study. Gynecol Endocrinol 2011; 27:1087-93. [PMID: 21190418 DOI: 10.3109/09513590.2010.540603] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Animal studies suggest regulatory effects on the hypothalamic-pituitary-gonad axis by allopregnanolone, an endogenous gamma-aminobutyric acid A (GABA(A)) receptor agonist. Elevated levels of allopregnanolone in women with hypothalamic amenorrhea have been seen. Isoallopregnanolone is an isomer to allopregnanolone, but without GABA(A) receptor effects. The purpose of this study was to investigate effects of allopregnanolone and isoallopregnanolone on gonadotropin levels in healthy women of fertile age. Ten women were given allopregnanolone and five women isoallopregnanolone intravenously in follicular phase. Repeated blood samples were drawn during the test day. Main outcomes were changes in serum levels of follicle-stimulating hormone (FSH), luteinising hormone (LH), oestradiol, and progesterone. Serum-FSH decreased between 5 and 105 min after the allopregnanolone injection (F(16,144)=2.18, p=0.008). Serum-LH was reduced between 5 and 35 min following the allopregnanolone injection (F(16,144)=2.63, p=0.001). Serum-oestradiol and -progesterone were not significantly changed after allopregnanolone injections. No effect on gonadotropin levels were seen after administration of isoallopregnanolone. Allopregnanolone reduces FSH and LH levels in women and the effect might be mediated via a specific GABA(A) receptor activation since isoallopregnanolone lacked this effect. Although the number of women was small, the results suggest a regulatory mechanism on the hypothalamic-pituitary-gonadal axis by allopregnanolon.
Collapse
Affiliation(s)
- Erika Timby
- Department of Clinical Science, Obstetrics and Gynaecology, Umeå Neurosteroid Research Centre, Umeå University, SE-901 85 Umeå, Sweden
| | | | | | | | | | | |
Collapse
|
26
|
Bäckström T, Haage D, Löfgren M, Johansson IM, Strömberg J, Nyberg S, Andréen L, Ossewaarde L, van Wingen GA, Turkmen S, Bengtsson SK. Paradoxical effects of GABA-A modulators may explain sex steroid induced negative mood symptoms in some persons. Neuroscience 2011; 191:46-54. [PMID: 21600269 DOI: 10.1016/j.neuroscience.2011.03.061] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2010] [Revised: 03/02/2011] [Accepted: 03/28/2011] [Indexed: 02/02/2023]
Abstract
Some women have negative mood symptoms, caused by progestagens in hormonal contraceptives or sequential hormone therapy or by progesterone in the luteal phase of the menstrual cycle, which may be attributed to metabolites acting on the GABA-A receptor. The GABA system is the major inhibitory system in the adult CNS and most positive modulators of the GABA-A receptor (benzodiazepines, barbiturates, alcohol, GABA steroids), induce inhibitory (e.g. anesthetic, sedative, anticonvulsant, anxiolytic) effects. However, some individuals have adverse effects (seizures, increased pain, anxiety, irritability, aggression) upon exposure. Positive GABA-A receptor modulators induce strong paradoxical effects including negative mood in 3%-8% of those exposed, while up to 25% have moderate symptoms. The effect is biphasic: low concentrations induce an adverse anxiogenic effect while higher concentrations decrease this effect and show inhibitory, calming properties. The prevalence of premenstrual dysphoric disorder (PMDD) is also 3%-8% among women in fertile ages, and up to 25% have more moderate symptoms of premenstrual syndrome (PMS). Patients with PMDD have severe luteal phase-related symptoms and show changes in GABA-A receptor sensitivity and GABA concentrations. Findings suggest that negative mood symptoms in women with PMDD are caused by the paradoxical effect of allopregnanolone mediated via the GABA-A receptor, which may be explained by one or more of three hypotheses regarding the paradoxical effect of GABA steroids on behavior: (1) under certain conditions, such as puberty, the relative fraction of certain GABA-A receptor subtypes may be altered, and at those subtypes the GABA steroids may act as negative modulators in contrast to their usual role as positive modulators; (2) in certain brain areas of vulnerable women the transmembrane Cl(-) gradient may be altered by factors such as estrogens that favor excitability; (3) inhibition of inhibitory neurons may promote disinhibition, and hence excitability. This article is part of a Special Issue entitled: Neuroactive Steroids: Focus on Human Brain.
Collapse
Affiliation(s)
- T Bäckström
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, SE-901 85 Umeå, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Johansson AGM, Nikamo P, Schalling M, Landén M. AKR1C4 gene variant associated with low euthymic serum progesterone and a history of mood irritability in males with bipolar disorder. J Affect Disord 2011; 133:346-51. [PMID: 21570127 DOI: 10.1016/j.jad.2011.04.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 04/08/2011] [Accepted: 04/08/2011] [Indexed: 01/27/2023]
Abstract
BACKGROUND Irritable mood during mood elevation is common in bipolar disorder. The progesterone metabolite allopregnanolone (ALLO) has been implicated in other disorders presenting with irritability. This study aimed to test whether a history of manic/hypomanic irritability is associated with low serum progesterone levels; and whether single nucleotide polymorphisms (SNPs) in gene coding for steroidogenetic enzymes (HSD3B2, SRD5A1 and AKR1C4 were coupled to previous manic irritability and/or with serum progesterone concentrations. METHODS Morning serum progesterone concentrations during euthymic phase of bipolar illness types 1 and 2 were assessed in 71 males and 107 females. Previous manic/hypomanic irritability was assessed using the Affective Disorders Evaluation. Selected SNPs were analyzed: i) aldoketoreductase-type-4 (AKR1C4 - rs17306779, rs3829125, rs10904440, rs12762017, and rs11253048), ii) 3-β-hydroxysteroid-dehydrogenase (HSD3B2 - rs4659174, rs2854964, and rs3765948), iii) steroid-5-α-reductase (SRD5A1 - rs8192139, rs181807, rs3822430, and rs3736316). RESULTS In males, progesterone concentrations were lower in those who had shown manic/hypomanic irritability compared with nonirritable (F=7.05, p=0.0099). SNPs rs17306779, rs3829125, and rs10904440 were associated with manic/hypomanic irritability. A cystine to serine change at position 145 in AKR1C4 (rs3829125) was associated with lower serum progesterone (F=6.34, p=0.014). There were no associations in females. LIMITATIONS Relatively small sample sizes. CONCLUSION Low progesterone levels and a cystine to serine change at position 145 in AKR1C4 gene are associated with manic/hypomanic irritability in males. Given that the enzyme AKR1C4 has both dehydrogenating and reductive activities in the steroidogenetic pathway, a missense variation in the gene may predispose to manic/hypomanic irritability by altering the relationship between progesterone and ALLO concentrations in the brain.
Collapse
Affiliation(s)
- Anette G M Johansson
- Division of Psychiatry, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | |
Collapse
|
28
|
Abstract
Neurosteroids represent a class of endogenous steroids that are synthesized in the brain, the adrenals, and the gonads and have potent and selective effects on the GABAA-receptor. 3α-hydroxy A-ring reduced metabolites of progesterone, deoxycorticosterone, and testosterone are positive modulators of GABA(A)-receptor in a non-genomic manner. Allopregnanolone (3α-OH-5α-pregnan-20-one), 5α-androstane-3α, 17α-diol (Adiol), and 3α5α-tetrahydrodeoxycorticosterone (3α5α-THDOC) enhance the GABA-mediated Cl(-) currents acting on a site (or sites) distinct from the GABA, benzodiazepine, barbiturate, and picrotoxin binding sites. 3α5α-P and 3α5α-THDOC potentiate synaptic GABA(A)-receptor function and activate δ-subunit containing extrasynaptic receptors that mediate tonic currents. On the contrary, 3β-OH pregnane steroids and pregnenolone sulfate (PS) are GABA(A)-receptor antagonists and induce activation-dependent inhibition of the receptor. The activities of neurosteroid are dependent on brain regions and types of neurons. In addition to the slow genomic action of the parent steroids, the non-genomic, and rapid actions of neurosteroids play a significant role in the GABA(A)-receptor function and shift in mood and memory function. This review describes molecular mechanisms underlying neurosteroid action on the GABA(A)-receptor, mood changes, and cognitive functions.
Collapse
Affiliation(s)
- Mingde Wang
- Section of Obstetrics and Gynecology, Department of Clinical Science, Umeå Neurosteroid Research Center, Umeå UniversityUmeå, Sweden
- *Correspondence: Mingde Wang, Section of Obstetrics and Gynecology, Department of Clinical Science, Umeå Neurosteroid Research Center, Umeå University, 901 85 Umeå, Sweden. e-mail:
| |
Collapse
|
29
|
Steckelbroeck S, Lütjohann D, Bauman DR, Ludwig M, Friedl A, Hans VHJ, Penning TM, Klingmüller D. Non-stereo-selective cytosolic human brain tissue 3-ketosteroid reductase is refractory to inhibition by AKR1C inhibitors. Biochim Biophys Acta Mol Cell Biol Lipids 2010; 1801:1221-31. [PMID: 20673851 DOI: 10.1016/j.bbalip.2010.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Revised: 07/16/2010] [Accepted: 07/19/2010] [Indexed: 01/02/2023]
Abstract
Cerebral 3α-hydroxysteroid dehydrogenase (3α-HSD) activity was suggested to be responsible for the local directed formation of neuroactive 5α,3α-tetrahydrosteroids (5α,3α-THSs) from 5α-dihydrosteroids. We show for the first time that within human brain tissue 5α-dihydroprogesterone and 5α-dihydrotestosterone are converted via non-stereo-selective 3-ketosteroid reductase activity to produce the respective 5α,3α-THSs and 5α,3β-THSs. Apart from this, we prove that within the human temporal lobe and limbic system cytochrome P450c17 and 3β-HSD/Δ(5-4) ketosteroid isomerase are not expressed. Thus, it appears that these brain regions are unable to conduct de novo biosynthesis of Δ(4)-3-ketosteroids from Δ(5)-3β-hydroxysteroids. Consequently, the local formation of THSs will depend on the uptake of circulating Δ(4)-3-ketosteroids such as progesterone and testosterone. 3α- and 3β-HSD activity were (i) equally enriched in the cytosol, (ii) showed equal distribution between cerebral neocortex and subcortical white matter without sex- or age-dependency, (iii) demonstrated a strong and significant positive correlation when comparing 46 different specimens and (iv) exhibited similar sensitivities to different inhibitors of enzyme activity. These findings led to the assumption that cerebral 3-ketosteroid reductase activity might be catalyzed by a single enzyme and is possibly attributed to the expression of a soluble AKR1C aldo-keto reductase. AKR1Cs are known to act as non-stereo-selective 3-ketosteroid reductases; low AKR1C mRNA expression was detected. However, the cerebral 3-ketosteroid reductase was clearly refractory to inhibition by AKR1C inhibitors indicating the expression of a currently unidentified enzyme. Its lack of stereo-selectivity is of physiological significance, since only 5α,3α-THSs enhance the effect of GABA on the GABA(A) receptor, whereas 5α,3β-THSs are antagonists.
Collapse
Affiliation(s)
- Stephan Steckelbroeck
- Institute of Clinical Chemistry and Pharmacology, University of Bonn, 53105 Bonn, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Andréen L, Nyberg S, Turkmen S, van Wingen G, Fernández G, Bäckström T. Sex steroid induced negative mood may be explained by the paradoxical effect mediated by GABAA modulators. Psychoneuroendocrinology 2009; 34:1121-1132. [PMID: 19272715 DOI: 10.1016/j.psyneuen.2009.02.003] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Revised: 02/05/2009] [Accepted: 02/06/2009] [Indexed: 10/21/2022]
Abstract
UNLABELLED Certain women experience negative mood symptoms as a result of progesterone during the luteal phase of the menstrual cycle, progestagens in hormonal contraceptives, or the addition of progesterone or progestagens in sequential hormone therapy (HT). This phenomenon is believed to be mediated via the action of the progesterone metabolites on the GABA(A) system, which is the major inhibitory system in the mammalian CNS. The positive modulators of the GABA(A) receptor include allopregnanolone and pregnanolone, both neuroactive metabolites of progesterone, as well as benzodiazepines, barbiturates, and alcohol. Studies on the effect of GABA(A) receptor modulators have shown contradictory results; although human and animal studies have revealed beneficial properties such as anaesthesia, sedation, anticonvulsant effects, and anxiolytic effects, recent reports have also indicated adverse effects such as anxiety, irritability, and aggression. It has actually been suggested that several GABA(A) receptor modulators, including allopregnanolone, have biphasic effects, in that low concentrations increase an adverse, anxiogenic effect whereas higher concentrations decrease this effect and show beneficial, calming properties. The allopregnanolone increase during the luteal phase in fertile women, as well as during the addition of progesterone in HT, has been shown to induce adverse mood in women. The severity of these mood symptoms is related to the allopregnanolone serum concentrations in a manner similar to an inverted U-shaped curve. Negative mood symptoms occur when the serum concentration of allopregnanolone is similar to endogenous luteal phase levels, while low and high concentrations have less effect on mood. It has also been shown that progesterone/allopregnanolone treatment in women increases the activity in the amygdala (as measured with functional magnetic resonance imaging) in a similar way to the changes seen during anxiety reactions. However, it is evident that only certain women experience adverse mood during progesterone or GABA(A) receptor modulator treatments. Women with premenstrual dysphoric disorder (PMDD) have severe luteal phase related symptoms; in this phase, they show changes in GABA(A) receptor sensitivity and GABA concentrations that are related to the severity of the condition. These findings suggest that negative mood symptoms in women with PMDD are caused by the paradoxical effect of allopregnanolone mediated via the GABA(A) receptor. CONCLUSION Progesterone and progestagens induce negative mood, most probably via their GABA(A) receptor active metabolites. In postmenopausal women treated with progesterone and animals treated with allopregnanolone, there is a bimodal association between serum allopregnanolone concentration and adverse mood, resembling an inverted U-shaped curve. In humans, the maximal effective concentration of allopregnanolone for producing negative mood is within the range of physiological luteal phase serum concentrations.
Collapse
Affiliation(s)
- Lotta Andréen
- Umeå Neurosteroid Research Center, Umeå University Department of Clinical Science, Obstetrics and Gynaecology, Norrlands University Hospital, Umeå SE-901 85, Sweden
| | | | | | | | | | | |
Collapse
|
31
|
Strömberg J, Lundgren P, Taube M, Bäckström T, Wang M, Haage D. The effect of the neuroactive steroid 5beta-pregnane-3beta, 20(R)-diol on the time course of GABA evoked currents is different to that of pregnenolone sulphate. Eur J Pharmacol 2009; 605:78-86. [PMID: 19168059 DOI: 10.1016/j.ejphar.2008.12.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2008] [Revised: 12/02/2008] [Accepted: 12/18/2008] [Indexed: 01/19/2023]
Abstract
The endogenous progesterone metabolite allopregnanolone has a number of properties including anesthetic, sedative, antiepileptic, anxiolytic, impaired memory function and negative mood symptoms. Allopregnanolone is a potent positive GABA(A) receptor function modulators. In contrast, 3beta-hydroxy-steroids (3beta-steroids) usually modulate the GABA(A) receptor negatively. They have attracted some interest for their possible use as therapeutic agents that could counteract the negative symptoms induced by allopregnanolone. Two hypotheses for the action of 3beta-steroids have been proposed: 1) 3beta-steroids act in a similar way to pregnenolone sulphate, which non-competitively reduces GABA(A) receptor activity. 2) 3beta-steroids specifically antagonize the effect of allopregnanolone. We have therefore tried to clarify this issue by comparing the effect of pregnenolone sulphate and 5beta-pregnane-3beta, 20(R)-diol on the GABA-evoked currents by the patch clamp technique on neurons from the medial preoptic nucleus. Both pregnenolone sulphate and 5beta-pregnane-3beta, 20(R)-diol increase the desensitization rate of the current response evoked by a 2 s GABA application. However, their effects on other parameters of the GABA evoked currents differed in degree and sometimes even in direction. The actions of pregnenolone sulphate and 5beta-pregnane-3beta, 20(R)-diol were not altered in the presence of allopregnanolone, which indicates that they do not directly interact with allopregnanolone. In addition, when 5beta-pregnane-3beta, 20(R)-diol was tested on spontaneous inhibitory postsynaptic currents (sIPSCs), it dramatically reduced the allopregnanolone-induced prolongation of the decay time constant but it had no effect on the decay under control conditions. In conclusion, the effect of 5beta-pregnane-3beta, 20(R)-diol on GABA-evoked currents is different to that of pregnenolone sulphate in medial preoptic nucleus neurons.
Collapse
Affiliation(s)
- Jessica Strömberg
- Department of Clinical Science, Umeå Neurosteroid Research Center, Umeå University, Norrlands University Hospital, Umeå, Sweden.
| | | | | | | | | | | |
Collapse
|
32
|
Hedström H, Bixo M, Nyberg S, Spigset O, Zingmark E, Bäckström T. Studies of pharmacokinetic and pharmacodynamic properties of isoallopregnanolone in healthy women. Psychopharmacology (Berl) 2009; 203:85-98. [PMID: 18949461 DOI: 10.1007/s00213-008-1372-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2008] [Accepted: 10/07/2008] [Indexed: 10/21/2022]
Abstract
RATIONALE The pharmacokinetics and behavioral effects of isoallopregnanolone (3beta-hydoxy-5alpha-pregnan-20-one) in women are not known. OBJECTIVES Allopregnanolone (3alpha-hydoxy-5alpha-pregnan-20-one) is a well-known neurosteroid, acting via the GABA(A) receptor in the human brain. The naturally occurring progesterone metabolite isoallopregnanolone is the 3beta-stereoisomer of allopregnanolone. Prior studies have concluded that isoallopregnanolone has no effect on the GABA(A) receptor. However, an antagonistic effect of isoallopregnanolone to allopregnanolone on the GABA(A) receptor has been shown in animal and in vitro studies. The purpose of this study was to evaluate the pharmacokinetics and behavioral effects of isoallopregnanolone in humans. MATERIALS AND METHODS Six healthy women were given three increasing doses of isoallopregnanolone intravenously in the follicular phase. Repeated blood samples for analyses of isoallopregnanolone and allopregnanolone concentrations were drawn. Saccadic eye movement variables, self-rated sedation, and mood rating scales were used during the test day. A Likert scale for prospective symptoms was used to measure daily fluctuations during the ongoing menstrual cycle. RESULTS Exogenously administered isoallopregnanolone produced a dose-dependent increase in the serum concentration of isoallopregnanolone. In parallel, there was also a rise in the allopregnanolone concentration. There was a decrease in saccadic eye movement variables, but no effect was found on self-rated sedation or mood and no changes were seen in prospective symptoms during the menstrual cycle. CONCLUSIONS After administration of isoallopregnanolone at a cumulative dose of 0.20 mg/kg, no adverse effects were observed. There is a metabolism of isoallopregnanolone to allopregnanolone, most likely explaining the effects on the saccadic eye movements.
Collapse
Affiliation(s)
- Helena Hedström
- Umeå Neurosteroid Research Center, Department of Clinical Science, Obstetrics, and Gynecology, Umeå University, Umeå, Sweden
| | | | | | | | | | | |
Collapse
|
33
|
Kask K, Bäckström T, Nilsson LG, Sundström-Poromaa I. Allopregnanolone impairs episodic memory in healthy women. Psychopharmacology (Berl) 2008; 199:161-168. [PMID: 18551282 DOI: 10.1007/s00213-008-1150-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2007] [Accepted: 03/17/2008] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Allopregnanolone is an endogenous neuroactive steroid that, through its binding to the gamma-aminobutyric acid (GABA) A receptor, has GABA-active properties. Animal studies indicate that allopregnanolone administration results in diminished learning and memory impairment. The aim of the current study was to investigate the effect of intravenously administered allopregnanolone on episodic memory, semantic memory, and working memory in healthy women. MATERIALS AND METHODS Twenty-eight healthy women were included in the study. The participants were scheduled for the memory tests twice in the follicular phase. During the test sessions, an intravenous allopregnanolone and placebo infusion were administered in a double-blinded, randomized order at intervals of 48 h. Before and 10 min after the allopregnanolone/placebo injections, memory tasks were performed. RESULTS The study demonstrated that allopregnanolone impaired episodic memory in healthy women. There was a significant difference between pre- and postallopregnanolone injection episodic memory scores (p < 0.05), whereas there was no change in episodic memory performance following the placebo injections. There was also a significant difference between allopregnanolone and placebo postinjection episodic memory scores (p < 0.05). There were no effects of allopregnanolone on the semantic memory task or working memory task. CONCLUSION Intravenous allopregnanolone impairs episodic memory in healthy women, but there is a high degree of individual variability.
Collapse
Affiliation(s)
- Kristiina Kask
- Department of Women's and Children's Health, University Hospital, Uppsala University, Uppsala, Sweden.
| | | | | | | |
Collapse
|
34
|
Wang MD, Borra VB, Strömberg J, Lundgren P, Haage D, Bäckström T. Neurosteroids 3beta, 20 (R/S)-pregnandiols decrease offset rate of the GABA-site activation at the recombinant GABA A receptor. Eur J Pharmacol 2008; 586:67-73. [PMID: 18374329 DOI: 10.1016/j.ejphar.2008.02.063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2007] [Revised: 02/07/2008] [Accepted: 02/20/2008] [Indexed: 10/22/2022]
Abstract
Neurosteroids directly modulate ligand gated ion channels such as GABA A receptors. Two such molecules, 3beta-OH A-ring reduced pregnane steroids and pregnenolone sulfate (PS), inhibit recombinant GABA A receptor. Using a two-electrode voltage-clamp technique, we compared the effect of 5alpha-pregnan-3beta,20(S)-diol (UC1019), 5beta-pregnan-3beta, 20(R)-diol (UC1020) and PS on the activation onset and offset times of the recombinant GABA A receptor (rat alpha1beta2gamma2L) in Xenopus oocytes. Rapid solution changes allowed the kinetic analysis of GABA-evoked currents. Steroids were co-applied with 30 microM GABA for 10 s, followed by a 80 s washout period. PS (> ir =0.3 microM) moderately increased the slow onset rate (k(on-S)) of GABA-response. PS had no significant effects on the fast onset rate (k(on-F)). UC1019 and UC1020 decreased the k(on-S) of the GABA-response in a concentration-dependent manner with no significant effects on the k(on-F). Like PS, UC1019 and UC1020 decreased the slow offset rates (k(off-S)). In addition, PS increased the fast offset rate (k(off-F)) in a concentration-dependent manner, while UC1019 and UC1020 decreased k(off-F). The EC50 of PS to increase k(off-F) was calculated as 0.47+/-0.1 microM. The corresponding IC50 values of UC1019 and UC1020 to decrease k(off-F) were 5.0+/-0.5 microM and 8.4+/-0.9 microM, respectively. These results suggest differential actions of PS and 3beta, 20(R/S)-pregnandiols on the offset time course of GABA-site activation.
Collapse
Affiliation(s)
- Ming-De Wang
- Umeå Neurosteroid Research Center, Department of Clinical Science, Obstetrics and Gynecology, Sweden.
| | | | | | | | | | | |
Collapse
|