1
|
Gao H, Sinha R, Wemm S, Milivojevic V. Pregnenolone effects on parasympathetic response to stress and alcohol cue provocation in treatment-seeking individuals with alcohol use disorder. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:619-628. [PMID: 39779217 PMCID: PMC11928267 DOI: 10.1111/acer.15529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Chronic alcohol consumption in alcohol use disorder (AUD) is associated with autonomic nervous system dysregulation, increasing cardiovascular risk, and high alcohol cravings. Heart rate variability (HRV), a marker of autonomic nervous system responsiveness to stressors, may mediate alcohol's impact on the cardiovascular system. While pregnenolone (PREG) has been shown to normalize heart rate and blood pressure in individuals with AUD, its effects on sympathetic and parasympathetic components of HRV and related alcohol craving are not known. METHODS Fifty-five treatment-seeking individuals with AUD were randomized to placebo (n = 21) or daily pregnenolone at 300 mg (n = 18) or 500 mg (n = 16), in a double-blind, 8-week pilot clinical trial. In week 2, participants underwent three randomized, counterbalanced 5-minute personalized guided imagery provocations (stress, alcohol, and neutral/relaxing cues) on separate days. HRV indices were assessed during each session and analyzed using linear mixed-effects models (LMEs), including association between HRV indices and anxiety and alcohol craving. RESULTS A medication group × condition interaction was found for parasympathetic, high-frequency (HF) (p = 0.028) and sympathetic/parasympathetic, low-frequency/high-frequency (LF/HF) ratio (p = 0.017) indices of HRV. Placebo had higher HF during alcohol cue (p = 0.011), while 500 mg PREG demonstrated lower HF in response to stress (p = 0.050) and alcohol cues (p = 0.047). Placebo showed lower LF/HF ratio during stress (p = 0.006) and alcohol cue (p = 0.001), while the PREG groups showed no changes. Overall, the LF/HF response to alcohol cue was significantly lower in placebo compared to the 300 mg PREG (p = 0.012) and 500 mg PREG (p = 0.037) groups. Lastly, HF was found to predict alcohol craving regardless of PREG doses. CONCLUSIONS We found a normalization of autonomic response in PREG groups. These findings suggest that PREG holds therapeutic potential for enhancing autonomic function in AUD.
Collapse
Affiliation(s)
- Huaze Gao
- The Yale Stress Center, Yale University School of Medicine, Department of Psychiatry, New Haven, CT 06519
| | - Rajita Sinha
- The Yale Stress Center, Yale University School of Medicine, Department of Psychiatry, New Haven, CT 06519
| | | | - Verica Milivojevic
- Address correspondence to: Verica Milivojevic, PhD, 2 Church Street South, Suite 209, New Haven, CT 06519, , Fax: 203-737-1272
| |
Collapse
|
2
|
Sakmar E, Wemm S, Fogelman N, Hermes G, Sinha R, Milivojevic V. Pregnenolone Reduces Provoked Craving and Cocaine Use in Men and Women with Cocaine Use Disorder: A Pilot Trial. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.07.25320141. [PMID: 39830237 PMCID: PMC11741489 DOI: 10.1101/2025.01.07.25320141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Aim Chronic cocaine use is associated with decreases in neuroactive steroid levels. These adaptations may contribute to continued cocaine use and high relapse risk in individuals with cocaine use disorder (CUD). Thus, this pilot study assessed chronic treatment with 2 supraphysiologic doses of the neuroactive steroid precursor pregnenolone (PREG, 300 mg/day; 500 mg/day) to boost endogenous neuroactive steroid levels and assess its impact on provoked craving and cocaine use outcomes in an 8-week trial in men and women with CUD. Methods Fifty-five treatment-seeking individuals with CUD were randomly assigned to receive either placebo (PLA; n=18; 12M/6F), 300mg PREG/day (n=20; 15M/5F) or 500mg PREG/day (n=17; 12M/5F) for 8 weeks, along with outpatient weekly relapse prevention treatment. Plasma was collected at weeks 2, 5 and 7 to assess circulating pregnenolone levels. A subset of subjects participated in a 3-day experimental component of guided imagery exposure to stress, cocaine cue and neutral conditions in about week 2 of the trial to assess craving response. Cocaine use outcomes was also assessed over the 8-week treatment period. Intent-to-treat analyses were conducted using linear mixed effects models. Results There were no differences between treatment groups on demographic variables and baseline cocaine use. Plasma pregnenolone levels were higher in the 300mg and 500mg PREG groups compared to PLA ( p's < 0.032). Participant trial completion rates were 100% for PLA, 90% for 300mg and 94% for 500mg PREG groups. Placebo group had increased craving in response to stress ( p < .001) and cocaine cue ( p < .001) provocation, whereas the PREG groups showed no increased in provoked cocaine craving. For cocaine use outcomes during the 8-week trial, a significant main effect of treatment group ( p = .005) on the weekly amounts of cocaine use showed e significantly lower amounts used in the 300mg PREG compared to the 500mg PREG group ( p = 0.01) and to PLA ( p = .047). There was also a trend for a treatment group main effect for days of cocaine used (p<.12). Conclusions These pilot findings suggest that supraphysiologic neuroactive steroid PREG doses reduces cocaine craving and may also improve cocaine use outcomes in treatment seeking individuals with CUD. Findings support further assessment and development of PREG in the treatment of CUD.
Collapse
|
3
|
Gao H, Magin Z, Fogelman N, Sinha R, Angarita GA, Milivojevic V. Stability and Reliability of Repeated Plasma Pregnenolone Levels After Oral Pregnenolone Dosing in Individuals with Cocaine Use Disorder: Pilot Findings. Life (Basel) 2024; 14:1483. [PMID: 39598281 PMCID: PMC11595496 DOI: 10.3390/life14111483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024] Open
Abstract
Substance use disorders (SUDs), including cocaine use disorder (CUD), have significant negative health risks and impose a substantial social burden, yet effective treatments are limited. Pregnenolone, a neuroactive steroid precursor, has been shown to reduce alcohol craving and normalize stress biology in individuals with CUD, but its clinical utility has been questioned due to limited data on bioavailability and the stability of blood levels in humans. Thus, this pilot study aimed to determine whether twice-daily oral pregnenolone (PREG) at 300 mg/day and 500 mg/day versus placebo in week two of PREG administration led to stable increased plasma pregnenolone levels in individuals with CUD. Seven treatment-seeking individuals with CUD, enrolled in an eight-week double-blind clinical trial, were randomized to receive placebo (n = 2) or pregnenolone at 300 mg/day (n = 3) or 500 mg/day (n = 2). For the first two weeks of the eight-week trial, participants were admitted to an inpatient Clinical Neuroscience Research Unit for repeated serial sampling of plasma pregnenolone concentrations over a 32.5 h period in week two of their inpatient stay while taking their assigned study drug under observation. Pregnenolone levels showed a significant main effect of the medication group (p = 0.039), with sustained higher levels in the 300 mg (p = 0.018) and 500 mg (p = 0.035) groups compared to placebo, and no significant difference between the two pregnenolone dosing groups. Moreover, correlation analyses showed that after observed study medication dosing on repeated sampling day 1, levels of pregnenolone were highly associated across time, with strong, positive correlations between time of dosing and 2 h (r = 0.80, p = 0.031), 4 h (r = 0.80, p = 0.031), 6 h (r = 0.86, p = 0.013), and 8 h post-dosing (r = 0.97, p < 0.001). These findings from this pilot study suggest that chronic twice-daily/"bis in die" (b.i.d.) oral administration of pregnenolone at both 300 mg/day and 500 mg/day achieved stable and reliable elevated plasma pregnenolone levels over 32.5 h in individuals with CUD, thereby supporting the good bioavailability of pregnenolone in these samples. These data indicate that twice-daily chronic dosing may overcome any potential concerns of poor bioavailability and rapid metabolism of pregnenolone in humans, and support further clinical investigations into pregnenolone's role in the treatment of cocaine use disorders.
Collapse
Affiliation(s)
- Huaze Gao
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA; (H.G.); (Z.M.); (N.F.); (R.S.)
| | - Zachary Magin
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA; (H.G.); (Z.M.); (N.F.); (R.S.)
| | - Nia Fogelman
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA; (H.G.); (Z.M.); (N.F.); (R.S.)
| | - Rajita Sinha
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA; (H.G.); (Z.M.); (N.F.); (R.S.)
| | - Gustavo A. Angarita
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, Yale University School of Medicine, New Haven, CT 06519, USA;
| | - Verica Milivojevic
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA; (H.G.); (Z.M.); (N.F.); (R.S.)
| |
Collapse
|
4
|
Chéry SL, O'Buckley TK, Boero G, Balan I, Morrow AL. Neurosteroid [3α,5α]3-hydroxypregnan-20-one inhibition of chemokine monocyte chemoattractant protein-1 in alcohol-preferring rat brain neurons, microglia, and astroglia. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1693-1703. [PMID: 38991981 DOI: 10.1111/acer.15404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND Neuroimmune dysfunction in alcohol use disorder (AUD) is associated with activation of myeloid differentiation primary response 88 (MyD88)-dependent Toll-like receptors (TLR) resulting in overexpression of the chemokine monocyte chemoattractant protein-1 (MCP-1/CCL2). MCP-1 overexpression in the brain is linked to anxiety, higher alcohol intake, neuronal death, and activation of microglia observed in AUD. The neurosteroid [3α,5α][3-hydroxypregnan-20-one (3α,5α-THP) has been reported as an inhibitor of MyD88-dependent TLR activation and MCP-1 overexpression in mouse and human macrophages and the brain of alcohol-preferring (P) rats. METHODS We investigated how 3α,5α-THP regulates MCP-1 expression at the cellular level in P rat nucleus accumbens (NAc) and central amygdala (CeA). We focused on neurons, microglia, and astrocytes, examining the individual voxel density of MCP-1, neuronal marker NeuN, microglial marker IBA1, astrocytic marker GFAP, and their shared voxel density, defined as intersection. Ethanol-naïve male and female P rats were perfused 1 h after IP injections of 15 mg/kg of 3α,5α-THP, or vehicle. The NAc and CeA were imaged using confocal microscopy following double-immunofluorescence staining for MCP-1 with NeuN, IBA1, and GFAP, respectively. RESULTS MCP-1 intersected with NeuN predominantly and IBA1/GFAP negligibly. 3α,5α-THP reduced MCP-1 expression in NeuN-labeled cells by 38.27 ± 28.09% in male and 56.11 ± 21.46% in female NAc, also 37.99 ± 19.53% in male and 54.96 ± 30.58% in female CeA. In females, 3α,5α-THP reduced the MCP-1 within IBA1 and GFAP-labeled voxels in the NAc and CeA. Conversely, in males, 3α,5α-THP did not significantly alter the MCP-1 within IBA1 in NAc or with GFAP in the CeA. Furthermore, 3α,5α-THP decreased levels of IBA1 in both regions and sexes with no impact on GFAP or NeuN levels. Secondary analysis performed on data normalized to % control values indicated that no significant sex differences were present. CONCLUSIONS These data suggest that 3α,5α-THP inhibits neuronal MCP-1 expression and decreases the proliferation of microglia in P rats. These results increase our understanding of potential mechanisms for 3α,5α-THP modulation of ethanol consumption.
Collapse
Affiliation(s)
- Samantha Lucenell Chéry
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Neuroscience Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Todd K O'Buckley
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Giorgia Boero
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pharmacology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Irina Balan
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - A Leslie Morrow
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pharmacology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
5
|
Maddern XJ, Ursich LT, Bailey G, Pearl A, Anversa RG, Lawrence AJ, Walker LC. Sex Differences in Alcohol Use: Is It All About Hormones? Endocrinology 2024; 165:bqae088. [PMID: 39018449 DOI: 10.1210/endocr/bqae088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/19/2024]
Abstract
Risky alcohol use and alcohol use disorders (AUD) are a rising problem in women, yet a major disparity in our understanding of what drives alcohol consumption in women remains. Historically biomedical research has focused on male subjects; however, recent increases in reporting of females, have highlighted major differences between the sexes. Here we review the current literature of the effect of gonadal steroid hormones (estrogens, androgens, and progestins), neurosteriods, and neurobiological factors on alcohol use in clinical and preclinical studies of both sexes. Further, we briefly discuss how fundamental sex differences in genetics, metabolism, neuroimmune, and stress responses may influence sex differences in alcohol intake. Comparing the sexes could aid in the discovery of novel therapeutics to treat AUD, and implementation of current treatment options in women.
Collapse
Affiliation(s)
- Xavier J Maddern
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Lauren T Ursich
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Grace Bailey
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Amy Pearl
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
| | - Roberta G Anversa
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| |
Collapse
|
6
|
Covault J, Tennen H, Feinn R. Randomized Placebo-Controlled Clinical Trial of Dutasteride for Reducing Heavy Drinking in Men. J Clin Psychopharmacol 2024; 44:223-231. [PMID: 38684046 PMCID: PMC11060692 DOI: 10.1097/jcp.0000000000001849] [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] [Indexed: 05/02/2024]
Abstract
BACKGROUND Prior studies indicate that neuroactive steroids mediate some of alcohol's effects. Dutasteride, widely used to treat benign prostatic hypertrophy, is an inhibitor of 5-alpha reductase enzymes, which play a central role in the production of 5α-reduced neuroactive steroids. The purpose of this study was to test dutasteride's tolerability and efficacy for reducing drinking. METHODS Men (n = 142) with heavy drinking (>24 drinks per week) and a goal to either stop or reduce drinking to nonhazardous levels were randomized to placebo or 1 mg dutasteride daily for 12 weeks. We hypothesized that dutasteride-treated patients would be more successful in reducing drinking. RESULTS Generalized linear mixed models that included baseline drinking, treatment, time and their 2-way interaction identified significant interactions of treatment-time, such that dutasteride treatment reduced drinking more than placebo. During the last month of treatment, 25% of dutasteride-treated participants had no hazardous drinking (no heavy drinking days and not more than 14 drinks per week) compared with 6% of placebo-treated participants (P = 0.006; NNT = 6). Sensitivity analysis identified baseline drinking to cope as a factor associated with larger reductions in drinking for dutasteride compared with placebo-treated participants. Dutasteride was well tolerated. Adverse events more common in the dutasteride group were stomach discomfort and reduced libido. CONCLUSION Dutasteride 1 mg daily was efficacious in reducing the number of heavy drinking days and drinks per week in treatment-seeking men. The benefit of dutasteride compared with placebo was greatest for participants with elevated baseline drinking to cope motives.
Collapse
Affiliation(s)
- Jonathan Covault
- Alcohol Research Center, Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT 06030
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269
| | - Howard Tennen
- Alcohol Research Center, Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT 06030
- Department of Public Health Sciences, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Richard Feinn
- Frank Netter School of Medicine, Quinnipiac University, Hamden, CT 06518
| |
Collapse
|
7
|
Johnston TH, Lacoste AMB, Ravenscroft P, Su J, Tamadon S, Seifi M, Lang AE, Fox SH, Brotchie JM, Visanji NP. Using artificial intelligence to identify drugs for repurposing to treat l-DOPA-induced dyskinesia. Neuropharmacology 2024; 248:109880. [PMID: 38412888 DOI: 10.1016/j.neuropharm.2024.109880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 02/02/2024] [Accepted: 02/18/2024] [Indexed: 02/29/2024]
Abstract
Repurposing regulatory agency-approved molecules, with proven safety in humans, is an attractive option for developing new treatments for disease. We identified and assessed the efficacy of 3 drugs predicted by an in silico screen as having the potential to treat l-DOPA-induced dyskinesia (LID) in Parkinson's disease. We analysed ∼1.3 million Medline abstracts using natural language processing and ranked 3539 existing drugs based on predicted ability to reduce LID. 3 drugs from the top 5% of the 3539 candidates; lorcaserin, acamprosate and ganaxolone, were prioritized for preclinical testing based on i) having a novel mechanism of action, ii) having not been previously validated for the treatment of LID, iii) being blood-brain-barrier penetrant and orally bioavailable and iv) being clinical trial ready. We assessed the efficacy of acamprosate, ganaxolone and lorcaserin in a rodent model of l-DOPA-induced hyperactivity, with lorcaserin affording a 58% reduction in rotational asymmetry (P < 0.05) compared to vehicle. Acamprosate and ganaxolone failed to demonstrate efficacy. Lorcaserin, a 5HT2C agonist, was then further tested in MPTP lesioned dyskinetic macaques where it afforded an 82% reduction in LID (P < 0.05), unfortunately accompanied by a significant increase in parkinsonian disability. In conclusion, although our data do not support the repurposing of lorcaserin, acamprosate or ganaxolone per se for LID, we demonstrate value of an in silico approach to identify candidate molecules which, in combination with an in vivo screen, can facilitate clinical development decisions. The present study adds to a growing literature in support of this paradigm shifting approach in the repurposing pipeline.
Collapse
Affiliation(s)
- Tom H Johnston
- Atuka Inc, Suite 5600, 100 King St. W. Toronto, Ontario, M5X 1C9, Canada
| | | | - Paula Ravenscroft
- Atuka Inc, Suite 5600, 100 King St. W. Toronto, Ontario, M5X 1C9, Canada
| | - Jin Su
- Atuka Inc, Suite 5600, 100 King St. W. Toronto, Ontario, M5X 1C9, Canada
| | - Sahar Tamadon
- Atuka Inc, Suite 5600, 100 King St. W. Toronto, Ontario, M5X 1C9, Canada
| | - Mahtab Seifi
- Atuka Inc, Suite 5600, 100 King St. W. Toronto, Ontario, M5X 1C9, Canada
| | - Anthony E Lang
- Krembil Brain Institute, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada; Edmond J Safra Program in Parkinson Disease, Parkinson Foundation Centre of Excellence, Toronto Western Hospital, 399, Bathurst St, Toronto, ON, M5T 2S8, Canada
| | - Susan H Fox
- Krembil Brain Institute, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada; Edmond J Safra Program in Parkinson Disease, Parkinson Foundation Centre of Excellence, Toronto Western Hospital, 399, Bathurst St, Toronto, ON, M5T 2S8, Canada
| | - Jonathan M Brotchie
- Krembil Brain Institute, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada; Atuka Inc, Suite 5600, 100 King St. W. Toronto, Ontario, M5X 1C9, Canada
| | - Naomi P Visanji
- Krembil Brain Institute, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada; Edmond J Safra Program in Parkinson Disease, Parkinson Foundation Centre of Excellence, Toronto Western Hospital, 399, Bathurst St, Toronto, ON, M5T 2S8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
8
|
Balan I, Boero G, Chéry SL, McFarland MH, Lopez AG, Morrow AL. Neuroactive Steroids, Toll-like Receptors, and Neuroimmune Regulation: Insights into Their Impact on Neuropsychiatric Disorders. Life (Basel) 2024; 14:582. [PMID: 38792602 PMCID: PMC11122352 DOI: 10.3390/life14050582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/18/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
Pregnane neuroactive steroids, notably allopregnanolone and pregnenolone, exhibit efficacy in mitigating inflammatory signals triggered by toll-like receptor (TLR) activation, thus attenuating the production of inflammatory factors. Clinical studies highlight their therapeutic potential, particularly in conditions like postpartum depression (PPD), where the FDA-approved compound brexanolone, an intravenous formulation of allopregnanolone, effectively suppresses TLR-mediated inflammatory pathways, predicting symptom improvement. Additionally, pregnane neurosteroids exhibit trophic and anti-inflammatory properties, stimulating the production of vital trophic proteins and anti-inflammatory factors. Androstane neuroactive steroids, including estrogens and androgens, along with dehydroepiandrosterone (DHEA), display diverse effects on TLR expression and activation. Notably, androstenediol (ADIOL), an androstane neurosteroid, emerges as a potent anti-inflammatory agent, promising for therapeutic interventions. The dysregulation of immune responses via TLR signaling alongside reduced levels of endogenous neurosteroids significantly contributes to symptom severity across various neuropsychiatric disorders. Neuroactive steroids, such as allopregnanolone, demonstrate efficacy in alleviating symptoms of various neuropsychiatric disorders and modulating neuroimmune responses, offering potential intervention avenues. This review emphasizes the significant therapeutic potential of neuroactive steroids in modulating TLR signaling pathways, particularly in addressing inflammatory processes associated with neuropsychiatric disorders. It advances our understanding of the complex interplay between neuroactive steroids and immune responses, paving the way for personalized treatment strategies tailored to individual needs and providing insights for future research aimed at unraveling the intricacies of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Irina Balan
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Giorgia Boero
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA;
| | - Samantha Lucenell Chéry
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Minna H. McFarland
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alejandro G. Lopez
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - A. Leslie Morrow
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
9
|
Peltier MR, Verplaetse TL, Altemus M, Zakiniaeiz Y, Ralevski EA, Mineur YS, Gueorguieva R, Picciotto MR, Cosgrove KP, Petrakis I, McKee SA. The role of neurosteroids in posttraumatic stress disorder and alcohol use disorder: A review of 10 years of clinical literature and treatment implications. Front Neuroendocrinol 2024; 73:101119. [PMID: 38184208 PMCID: PMC11185997 DOI: 10.1016/j.yfrne.2023.101119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 12/08/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
Rates of alcohol use disorder (AUD) are increasing in men and women and there are high rates of concurrent posttraumatic stress disorder (PTSD) and AUD. AUD and PTSD synergistically increase symptomatology and negatively affect treatment outcomes; however, there are very limited pharmacological treatments for PTSD/AUD. Neurosteroids have been implicated in the underlying neurobiological mechanisms of both PTSD and AUD and may be a target for treatment development. This review details the past ten years of research on pregnenolone, progesterone, allopregnanolone, pregnanolone, estradiol, testosterone and dehydroepiandrosterone/dehydroepiandrosterone-sulfate (DHEA/DHEA-S) in the context of PTSD and AUD, including examination of trauma/alcohol-related variables, such as stress-reactivity. Emerging evidence that exogenous pregnenolone, progesterone, and allopregnanolone may be promising, novel interventions is also discussed. Specific emphasis is placed on examining the application of sex as a biological variable in this body of literature, given that women are more susceptible to both PTSD diagnoses and stress-related alcohol consumption.
Collapse
Affiliation(s)
- MacKenzie R Peltier
- Yale School of Medicine, Department of Psychiatry, New Haven, CT 06519, USA; VA Connecticut Healthcare System, Mental Health Service, West Haven, CT 06516, USA; National Center for PTSD, Clinical Neuroscience Division, West Haven, CT 06516, USA.
| | | | - Margaret Altemus
- Yale School of Medicine, Department of Psychiatry, New Haven, CT 06519, USA; VA Connecticut Healthcare System, Mental Health Service, West Haven, CT 06516, USA
| | - Yasmin Zakiniaeiz
- Yale School of Medicine, Department of Psychiatry, New Haven, CT 06519, USA
| | - Elizabeth A Ralevski
- Yale School of Medicine, Department of Psychiatry, New Haven, CT 06519, USA; VA Connecticut Healthcare System, Mental Health Service, West Haven, CT 06516, USA
| | - Yann S Mineur
- Yale School of Medicine, Department of Psychiatry, New Haven, CT 06519, USA
| | - Ralitza Gueorguieva
- Yale School of Medicine, Department of Psychiatry, New Haven, CT 06519, USA; Department of Biostatistics, School of Public Health, Yale University, New Haven, CT, USA
| | - Marina R Picciotto
- Yale School of Medicine, Department of Psychiatry, New Haven, CT 06519, USA
| | - Kelly P Cosgrove
- Yale School of Medicine, Department of Psychiatry, New Haven, CT 06519, USA; National Center for PTSD, Clinical Neuroscience Division, West Haven, CT 06516, USA; Department of Radiology and Biomedical Imaging, School of Medicine, Yale University, New Haven, CT, USA
| | - Ismene Petrakis
- Yale School of Medicine, Department of Psychiatry, New Haven, CT 06519, USA; VA Connecticut Healthcare System, Mental Health Service, West Haven, CT 06516, USA; National Center for PTSD, Clinical Neuroscience Division, West Haven, CT 06516, USA
| | - Sherry A McKee
- Yale School of Medicine, Department of Psychiatry, New Haven, CT 06519, USA
| |
Collapse
|
10
|
Belelli D, Riva A, Nutt DJ. Reducing the harms of alcohol: nutritional interventions and functional alcohol alternatives. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 175:241-276. [PMID: 38555118 DOI: 10.1016/bs.irn.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
The health risks and harm associated with regular alcohol consumption are well documented. In a recent WHO statement published in The Lancet Public Health alcohol consumption has been estimated to contribute worldwide to 3 million deaths in 2016 while also being responsible for 5·1% of the global burden of disease and injury. The total elimination of alcohol consumption, which has been long imbedded in human culture and society, is not practical and prohibition policies have proved historically ineffective. However, valuable strategies to reduce alcohol harms are already available and improved alternative approaches are currently being developed. Here, we will review and discuss recent advances on two main types of approaches, that is nutritional interventions and functional alcohol alternatives.
Collapse
Affiliation(s)
- Delia Belelli
- GABALabs Res. Senior Scientific Consultant, United Kingdom
| | - Antonio Riva
- Roger Williams Institute of Hepatology (Foundation for Liver Research), London; Faculty of Life Sciences & Medicine, King's College London, London
| | | |
Collapse
|
11
|
Morrow AL, Boero G, Balan I. Emerging evidence for endogenous neurosteroid modulation of pro-inflammatory and anti-inflammatory pathways that impact neuropsychiatric disease. Neurosci Biobehav Rev 2024; 158:105558. [PMID: 38244954 DOI: 10.1016/j.neubiorev.2024.105558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/01/2023] [Accepted: 01/16/2024] [Indexed: 01/22/2024]
Abstract
This mini-review presents emerging evidence that endogenous neurosteroids modulate both pro- and anti-inflammatory signaling by immune cells and brain cells that contribute to depression, alcohol use disorders, and other inflammatory conditions. We first review the literature on pregnenolone and allopregnanolone inhibition of proinflammatory neuroimmune pathways in the periphery and the brain - effects that are independent of GABAergic mechanisms. We follow with evidence for neurosteroid enhancement of anti-inflammatory and protective pathways in brain and immune cells. These studies draw clinical relevance from a large body of evidence that pro-inflammatory immune signaling is dysregulated in many brain disorders and the fact that neurosteroids inhibit the same inflammatory pathways that are activated in depression, alcohol use disorders and other inflammatory conditions. Thus, we describe evidence that neurosteroid levels are decreased and neurosteroid supplementation has therapeutic efficacy in these neuropsychiatric conditions. We conclude with a perspective that endogenous regulation of immune balance between pro- and anti-inflammatory pathways by neurosteroid signaling is essential to prevent the onset of disease. Deficits in neurosteroids may unleash excessive pro-inflammatory activation which progresses in a feed-forward manner to disrupt brain networks that regulate stress, emotion and motivation. Neurosteroids can block various inflammatory pathways in mouse and human macrophages, rat brain and human blood and therefore provide new hope for treatment of intractable conditions that involve excessive inflammatory signaling.
Collapse
Affiliation(s)
- A Leslie Morrow
- Department of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Giorgia Boero
- Department of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Irina Balan
- Department of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
12
|
Finn DA. Stress and gonadal steroid influences on alcohol drinking and withdrawal, with focus on animal models in females. Front Neuroendocrinol 2023; 71:101094. [PMID: 37558184 PMCID: PMC10840953 DOI: 10.1016/j.yfrne.2023.101094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/06/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023]
Abstract
Sexually dimorphic effects of alcohol, following binge drinking, chronic intoxication, and withdrawal, are documented at the level of the transcriptome and in behavioral and physiological responses. The purpose of the current review is to update and to expand upon contributions of the endocrine system to alcohol drinking and withdrawal in females, with a focus on animal models. Steroids important in the hypothalamic-pituitary-gonadal and hypothalamic-pituitary-adrenal axes, the reciprocal interactions between these axes, the effects of chronic alcohol use on steroid levels, and the genomic and rapid membrane-associated effects of steroids and neurosteroids in models of alcohol drinking and withdrawal are described. Importantly, comparison between males and females highlight some divergent effects of sex- and stress-steroids on alcohol drinking- and withdrawal-related behaviors, and the distinct differences in response emphasize the importance of considering sex in the development of novel pharmacotherapies for the treatment of alcohol use disorder.
Collapse
Affiliation(s)
- Deborah A Finn
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States; Department of Research, VA Portland Health Care System, Portland, OR, United States.
| |
Collapse
|
13
|
Boero G, McFarland MH, Tyler RE, O’Buckley TK, Chéry SL, Robinson DL, Besheer J, Morrow AL. Deleterious Interaction between the Neurosteroid (3α,5α)3-Hydroxypregnan-20-One (3α,5α-THP) and the Mu-Opioid System Activation during Forced Swim Stress in Rats. Biomolecules 2023; 13:1205. [PMID: 37627270 PMCID: PMC10452864 DOI: 10.3390/biom13081205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
The neurosteroid 3α,5α-THP is a potent GABAA receptor-positive modulator and its regulatory action on the HPA axis stress response has been reported in numerous preclinical and clinical studies. We previously demonstrated that 3α,5α-THP down-regulation of HPA axis activity during stress is sex-, brain region- and stressor-dependent. In this study, we observed a deleterious submersion behavior in response to 3α,5α-THP (15 mg/kg) during forced swim stress (FSS) that led us to investigate how 3α,5α-THP might affect behavioral coping strategies engaged in by the animal. Given the well-established involvement of the opioid system in HPA axis activation and its interaction with GABAergic neurosteroids, we explored the synergic effects of 3α,5α-THP/opiate system activation in this behavior. Serum β-endorphin (β-EP) was elevated by FSS and enhanced by 3α,5α-THP + FSS. Hypothalamic Mu-opiate receptors (MOP) were increased in female rats by 3α,5α-THP + FSS. Pretreatment with the MOP antagonist D-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2 (CTAP; 2 mg/kg, IP) reversed submersion behavior in males. Moreover, in both males and females, CTAP pretreatment decreased immobility episodes while increasing immobility duration but did not alter swimming duration. This interaction between 3α,5α-THP and the opioid system in the context of FSS might be important in the development of treatment for neuropsychiatric disorders involving HPA axis activation.
Collapse
Affiliation(s)
- Giorgia Boero
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Bldg., CB 7178, Chapel Hill, NC 27599, USA; (G.B.)
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Minna H. McFarland
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Bldg., CB 7178, Chapel Hill, NC 27599, USA; (G.B.)
| | - Ryan E. Tyler
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Bldg., CB 7178, Chapel Hill, NC 27599, USA; (G.B.)
| | - Todd K. O’Buckley
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Bldg., CB 7178, Chapel Hill, NC 27599, USA; (G.B.)
| | - Samantha L. Chéry
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Bldg., CB 7178, Chapel Hill, NC 27599, USA; (G.B.)
| | - Donita L. Robinson
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Bldg., CB 7178, Chapel Hill, NC 27599, USA; (G.B.)
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Joyce Besheer
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Bldg., CB 7178, Chapel Hill, NC 27599, USA; (G.B.)
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - A. Leslie Morrow
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, 3027 Thurston Bowles Bldg., CB 7178, Chapel Hill, NC 27599, USA; (G.B.)
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
14
|
Ornelas LC, Boero G, Van Voorhies K, O’Buckley TK, Besheer J, Morrow AL. Pharmacological administration of 3α,5α-THP into the nucleus accumbens core increases 3α,5α-THP expression and reduces alcohol self-administration. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:459-469. [PMID: 36587947 PMCID: PMC10234128 DOI: 10.1111/acer.15008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/16/2022] [Accepted: 12/23/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND Alcohol affects multiple circuits in the brain, mainly disrupting the delicate balance between inhibitory γ-aminobutyric acid (GABA) transmission and excitatory glutamate signaling in brain areas involved in reward circuits. These include the amygdala, nucleus accumbens (Acb), and ventral tegmental area (VTA). This action impairs circuits that regulate behavioral control of craving and alcohol seeking and intake. Studies in both rodent models and postmortem human brain of patients with alcohol use disorder (AUD) have highlighted the association between the loss of GABAergic inhibition and the development of addiction. The neurosteroid (3α,5α)-3-hydroxypregnan-20-one (3α,5α-THP) is a potent positive modulator of GABAA receptors. Chronic alcohol consumption reduces 3α,5α-THP levels, resulting in decreased GABA inhibition. We previously demonstrated that enhancing neurosteroid biosynthesis by overexpression of the cholesterol side-chain cleavage enzyme P450scc decreased alcohol intake in male alcohol-preferring rats (P-rats). While most of the evidence of alcohol-induced alterations comes from studies in male subjects, some data show that females are more vulnerable to alcohol's effects than males. METHODS In this study, we investigated the ability of 3α,5α-THP direct infusions in two brain regions that contribute to alcohol reinforcement, the VTA and Acb core (AcbC), to regulate alcohol self-administration in female P-rats. RESULTS Administration of 3α,5α-THP into the AcbC increased 3α,5α-THP-positive cell expression in this area and reduced alcohol self-administration. By contrast, 3α,5α-THP infusion into the VTA did not significantly affect alcohol self-administration, though trends for a reduction were found. CONCLUSIONS Our results show that local increases in 3α,5α-THP in the AcbC may alter mesolimbic activity that drives a reduction in alcohol self-administration.
Collapse
Affiliation(s)
- Laura C. Ornelas
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Giorgia Boero
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Kalynn Van Voorhies
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Todd K. O’Buckley
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Joyce Besheer
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - A. Leslie Morrow
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| |
Collapse
|
15
|
Frau R, Melis M. Sex-specific susceptibility to psychotic-like states provoked by prenatal THC exposure: Reversal by pregnenolone. J Neuroendocrinol 2023; 35:e13240. [PMID: 36810840 DOI: 10.1111/jne.13240] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/23/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
Sociocultural attitudes towards cannabis legalization contribute to the common misconception that it is a relatively safe drug and its use during pregnancy poses no risk to the fetus. However, longitudinal studies demonstrate that maternal cannabis exposure results in adverse outcomes in the offspring, with a heightened risk for developing psychopathology. One of the most reported psychiatric outcomes is the proneness to psychotic-like experiences during childhood. How exposure to cannabis during gestation increases psychosis susceptibility in children and adolescents remains elusive. Preclinical research has indicated that in utero exposure to the major psychoactive component of cannabis, delta-9-tetrahydrocannabinol (THC), deranges brain developmental trajectories towards vulnerable psychotic-like endophenotypes later in life. Here, we present how prenatal THC exposure (PCE) deregulates mesolimbic dopamine development predisposing the offspring to schizophrenia-relevant phenotypes, exclusively when exposed to environmental challenges, such as stress or THC. Detrimental effects of PCE are sex-specific because female offspring do not display psychotic-like outcomes upon exposure to these challenges. Moreover, we present how pregnenolone, a neurosteroid that showed beneficial properties on the effects elicited by cannabis intoxication, normalizes mesolimbic dopamine function and rescues psychotic-like phenotypes. We, therefore, suggest this neurosteroid as a safe "disease-modifying" aid to prevent the onset of psychoses in vulnerable individuals. Our findings corroborate clinical evidence and highlight the relevance of early diagnostic screening and preventative strategies for young individuals at risk for mental diseases, such as male PCE offspring.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
- The Guy Everett Laboratory for Neuroscience, University of Cagliari, Cagliari, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
| |
Collapse
|
16
|
Milivojevic V, Sullivan L, Tiber J, Fogelman N, Simpson C, Hermes G, Sinha R. Pregnenolone effects on provoked alcohol craving, anxiety, HPA axis, and autonomic arousal in individuals with alcohol use disorder. Psychopharmacology (Berl) 2023; 240:101-114. [PMID: 36445398 PMCID: PMC10630889 DOI: 10.1007/s00213-022-06278-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 11/10/2022] [Indexed: 11/30/2022]
Abstract
RATIONALE Chronic alcohol intake down-regulates GABAergic transmission and reduces levels of neuroactive steroids. These changes are associated with greater stress dysregulation and high alcohol craving which in turn increases relapse risk. OBJECTIVES This study tested whether potentiation of the neurosteroid system with pregnenolone (PREG), a precursor to neuroactive steroids and known to increase GABAergic transmission, will normalize chronic alcohol-related stress adaptations in the hypothalamic-pituitary-adrenal (HPA) axis and autonomic responses and reduce alcohol craving to significantly impact relapse risk. METHODS Forty-three treatment-seeking individuals with alcohol use disorder (AUD) were randomized to placebo (PBO) or supraphysiologic pregnenolone doses of 300 mg or 500 mg treatment using a parallel-between subject design as part of a larger 8-week pilot clinical trial. In week 2, they participated in a 3-day laboratory experiment where on each day they self-administered the assigned study drug in the laboratory and were then exposed to 5-min personalized guided imagery provocation of stress, alcohol, or neutral/relaxing cues, one condition per day on separate days, in a random, counterbalanced order. Repeated assessments of alcohol craving, anxiety, HPA axis, heart rate (HR), systolic (SBP), and diastolic blood pressure (DBP) and serum pregnenolone levels were made on each day. RESULTS Pregnenolone levels were significantly increased in the PREG groups versus PBO. PREG treatment decreased stress- and alcohol cue- induced craving and dose-specifically reduced stress-induced anxiety in the 300 mg/day group. Both PREG doses compared to PBO also normalized CORT/ACTH and increased stress-induced HR, stress- and cue-induced SBP, and in the 300 mg PREG group cue-induced DBP responses relative to neutral condition. CONCLUSIONS Findings indicate that pregnenolone decreases stress- and alcohol cue-provoked craving and normalizes HPA axis and autonomic arousal in individuals with AUD, thereby supporting the need for further assessment of pregnenolone in the treatment of AUD.
Collapse
Affiliation(s)
- Verica Milivojevic
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA.
| | - Liam Sullivan
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Jessica Tiber
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Nia Fogelman
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Christine Simpson
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Gretchen Hermes
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Rajita Sinha
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| |
Collapse
|
17
|
Milivojevic V, Charron L, Fogelman N, Hermes G, Sinha R. Pregnenolone Reduces Stress-Induced Craving, Anxiety, and Autonomic Arousal in Individuals with Cocaine Use Disorder. Biomolecules 2022; 12:biom12111593. [PMID: 36358943 PMCID: PMC9687893 DOI: 10.3390/biom12111593] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic cocaine use leads to adaptations in stress biology and in neuroactive steroid system. These adaptations are associated with high cocaine craving and increased relapse risk. This study tested whether potentiation of the neuroactive steroid system with the precursor pregnenolone (PREG) affects stress- and cue-induced cocaine craving, anxiety and autonomic response in individuals with cocaine use disorder (CUD). Thirty treatment-seeking individuals (21 Male, 9 Female) with CUD were randomized to placebo (PBO) or supraphysiologic PREG doses of 300 mg or 500 mg per day for 8 weeks. After 2 weeks of treatment, participants were exposed to 5-min personalized guided imagery provocation of stress, cocaine, or neutral/relaxing cues in a 3-day experiment, one condition per day on separate days, in a random, counterbalanced order. Repeated assessment of cocaine craving, anxiety, heart rate (HR), systolic (SBP) and diastolic blood pressure (DBP) were assessed on each day. PREG significantly increased pregnenolone levels compared to PBO. Both PREG doses decreased stress- and cocaine cue-induced craving and reduced both stress- and cue-induced anxiety only in the 500 mg/day group. The 500 mg/day PREG group also displayed decreased stress-induced HR, SBP and DBP. Findings indicate that pregnenolone decreases stress- and cocaine cue-provoked craving and anxiety and reduces stress-induced autonomic arousal in individuals with CUD.
Collapse
|
18
|
Lattanzi S, Riva A, Striano P. Ganaxolone treatment for epilepsy patients: from pharmacology to place in therapy. Expert Rev Neurother 2021; 21:1317-1332. [PMID: 33724128 DOI: 10.1080/14737175.2021.1904895] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/15/2021] [Indexed: 02/08/2023]
Abstract
Introduction: Nonsulfated neurosteroids can provide phasic and tonic inhibition through activation of synaptic and extra-synaptic γ-aminobutyric acid (GABA)A receptors, exhibiting a greater potency for the latter. These actions occur by interacting with modulatory sites that are distinct from those bound by benzodiazepines and barbiturates. Ganaxolone (GNX) is a synthetic analog of the endogenous neurosteroid allopregnanolone and a member of a novel class of neuroactive steroids called epalons.Areas covered: The authors review the pharmacology of GNX, summarize the main clinical evidence about its antiseizure efficacy and tolerability, and suggest implications for clinical practice and future research.Expert opinion: The clinical development of GNX is mainly oriented to target unmet needs and focused on status epilepticus and rare genetic epilepsies that have few or no treatment options.The availability of oral and intravenous formulations allows reaching adult and pediatric patients in acute and chronic care settings. Further evidence will complement the understanding of the potentialities of GNX and possibly lead to indications for use in clinical practice.
Collapse
Affiliation(s)
- Simona Lattanzi
- Neurological Clinic, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Antonella Riva
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, "G. Gaslini" Institute, University of Genoa, Genova, Italy
| | - Pasquale Striano
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, "G. Gaslini" Institute, University of Genoa, Genova, Italy
| |
Collapse
|
19
|
El-Mallakh RS, Ali Z. Extra-synaptic modulation of GABA A and efficacy in bipolar disorder. Med Hypotheses 2021; 147:110501. [PMID: 33515862 DOI: 10.1016/j.mehy.2021.110501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/22/2020] [Accepted: 01/08/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Bipolar disorder type I is a severe psychiatric condition that leads to significant morbidity and mortality and whose treatment remains suboptimal. Its pathophysiology involves disturbance in the control of ionic fluxes so that when patients are either manic or depressed, the resting membrane potential of neurons is more depolarized than normal. Available mood stabilizers have a shared mechanism of normalizing ion flux by compensating for ionic abnormalities, and normalizing membrane potential. HYPOTHESIS Agents that significantly potentiate extrasynaptic GABAA receptors are expected to be particularly effective in hyperpolarizing resting membrane potential in bipolar patients, thereby normalizing their membrane potential. DISCUSSION New neuroactive steroid-like agents are being tested in humans for depression and insomnia. These agents include brexanolone, ganaxolone, and gaboxadol. Brexanolone has been approved for the treatment of postpartum depression, ganaxolone is being studied for treatment-resistant depression, and gaboxadol development for the treatment of insomnia has been abandoned due to narrow therapeutic index. In addition to the current studies, these agents are expected to have particular efficacy in acute and prophylactic management of bipolar I disorder by hyperpolarizing the resting potential of neurons and antagonizing one of the most reproducible demonstrated biologic abnormalities of this illness.
Collapse
Affiliation(s)
- Rif S El-Mallakh
- Mood Disorders Research Program, Depression Center Department of Psychiatry and Behavioral Sciences University of Louisville School of Medicine, 401 East Chestnut Street, Suite 610 Louisville, Kentucky 40202, USA.
| | - Ziad Ali
- Department of Psychiatry, University of Kentucky College of Medicine, The Medical Center of Bowling Green, Bowling Green, KY, USA
| |
Collapse
|
20
|
Abstract
Sexually dimorphic effects of alcohol exposure throughout life have been documented in clinical and preclinical studies. In the past, rates of alcohol use disorder (AUD) were higher in men than in women, but over the past 10 years, the difference between sexes in prevalence of AUD and binge drinking has narrowed. Recent evidence adds to historical data regarding the influence of sex steroids on alcohol drinking and the interaction with stress-related steroids. This review considers the contribution of the endocrine system to alcohol drinking in females, with a focus on the hypothalamic pituitary gonadal axis and the hypothalamic pituitary adrenal axis and their reciprocal interactions. Emphasis is given to preclinical studies that examined genomic and rapid membrane effects of estrogen, progesterone, glucocorticoids, and GABAergic neurosteroids for their effects on alcohol drinking and models of relapse. Pertinent comparisons to data in males highlight divergent effects of sex and stress steroids on alcohol drinking and emphasize the importance of considering sex in the development of novel pharmacotherapeutic targets for the treatment of AUD. For instance, pharmacological strategies targeting the corticotropin releasing factor and glucocorticoid receptor systems may be differentially effective in males and females, whereas strategies to enhance GABAergic neurosteroids may represent a biomarker of treatment efficacy in both sexes.
Collapse
Affiliation(s)
- Deborah A Finn
- Oregon Health & Science University, Portland, Oregon.,Veterans Affairs Portland Health Care System, Portland, Oregon
| |
Collapse
|
21
|
Morrow AL, Boero G, Porcu P. A Rationale for Allopregnanolone Treatment of Alcohol Use Disorders: Basic and Clinical Studies. Alcohol Clin Exp Res 2020; 44:320-339. [PMID: 31782169 PMCID: PMC7018555 DOI: 10.1111/acer.14253] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 11/19/2019] [Indexed: 12/17/2022]
Abstract
For many years, research from around the world has suggested that the neuroactive steroid (3α,5α)-3-hydroxypregnan-20-one (allopregnanolone or 3α,5α-THP) may have therapeutic potential for treatment of various symptoms of alcohol use disorders (AUDs). In this critical review, we systematically address all the evidence that supports such a suggestion, delineate the etiologies of AUDs that are addressed by treatment with allopregnanolone or its precursor pregnenolone, and the rationale for treatment of various components of the disease based on basic science and clinical evidence. This review presents a theoretical framework for understanding how endogenous steroids that regulate the effects of stress, alcohol, and the innate immune system could play a key role in both the prevention and the treatment of AUDs. We further discuss cautions and limitations of allopregnanolone or pregnenolone therapy with suggestions regarding the management of risk and the potential for helping millions who suffer from AUDs.
Collapse
Affiliation(s)
- A. Leslie Morrow
- Department of Psychiatry, Department of Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599
| | - Giorgia Boero
- Department of Psychiatry, Department of Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599
| | - Patrizia Porcu
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| |
Collapse
|
22
|
Ferguson LB, Patil S, Moskowitz BA, Ponomarev I, Harris RA, Mayfield RD, Messing RO. A Pathway-Based Genomic Approach to Identify Medications: Application to Alcohol Use Disorder. Brain Sci 2019; 9:E381. [PMID: 31888299 PMCID: PMC6956180 DOI: 10.3390/brainsci9120381] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/31/2022] Open
Abstract
Chronic, excessive alcohol use alters brain gene expression patterns, which could be important for initiating, maintaining, or progressing the addicted state. It has been proposed that pharmaceuticals with opposing effects on gene expression could treat alcohol use disorder (AUD). Computational strategies comparing gene expression signatures of disease to those of pharmaceuticals show promise for nominating novel treatments. We reasoned that it may be sufficient for a treatment to target the biological pathway rather than lists of individual genes perturbed by AUD. We analyzed published and unpublished transcriptomic data using gene set enrichment of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways to identify biological pathways disrupted in AUD brain and by compounds in the Library of Network-based Cellular Signatures (LINCS L1000) and Connectivity Map (CMap) databases. Several pathways were consistently disrupted in AUD brain, including an up-regulation of genes within the Complement and Coagulation Cascade, Focal Adhesion, Systemic Lupus Erythematosus, and MAPK signaling, and a down-regulation of genes within the Oxidative Phosphorylation pathway, strengthening evidence for their importance in AUD. Over 200 compounds targeted genes within those pathways in an opposing manner, more than twenty of which have already been shown to affect alcohol consumption, providing confidence in our approach. We created a user-friendly web-interface that researchers can use to identify drugs that target pathways of interest or nominate mechanism of action for drugs. This study demonstrates a unique systems pharmacology approach that can nominate pharmaceuticals that target pathways disrupted in disease states such as AUD and identify compounds that could be repurposed for AUD if sufficient evidence is attained in preclinical studies.
Collapse
Affiliation(s)
- Laura B. Ferguson
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; (L.B.F.); (S.P.); (B.A.M.); (R.A.H.); (R.D.M.)
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Shruti Patil
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; (L.B.F.); (S.P.); (B.A.M.); (R.A.H.); (R.D.M.)
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Bailey A. Moskowitz
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; (L.B.F.); (S.P.); (B.A.M.); (R.A.H.); (R.D.M.)
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Igor Ponomarev
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Robert A. Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; (L.B.F.); (S.P.); (B.A.M.); (R.A.H.); (R.D.M.)
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Roy D. Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; (L.B.F.); (S.P.); (B.A.M.); (R.A.H.); (R.D.M.)
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Robert O. Messing
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA; (L.B.F.); (S.P.); (B.A.M.); (R.A.H.); (R.D.M.)
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
23
|
Tomaselli G, Vallée M. Stress and drug abuse-related disorders: The promising therapeutic value of neurosteroids focus on pregnenolone-progesterone-allopregnanolone pathway. Front Neuroendocrinol 2019; 55:100789. [PMID: 31525393 DOI: 10.1016/j.yfrne.2019.100789] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/14/2019] [Accepted: 09/09/2019] [Indexed: 02/06/2023]
Abstract
The pregnenolone-progesterone-allopregnanolone pathway is receiving increasing attention in research on the role of neurosteroids in pathophysiology, particularly in stress-related and drug use disorders. These disorders involve an allostatic change that may result from deficiencies in allostasis or adaptive responses, and may be downregulated by adjustments in neurotransmission by neurosteroids. The following is an overview of findings that assess how pregnenolone and/or allopregnanolone concentrations are altered in animal models of stress and after consumption of alcohol or cannabis-type drugs, as well as in patients with depression, anxiety, post-traumatic stress disorder or psychosis and/or in those diagnosed with alcohol or cannabis use disorders. Preclinical and clinical evidence shows that pregnenolone and allopregnanolone, operating according to a different or common pharmacological profile involving GABAergic and/or endocannabinoid system, may be relevant biomarkers of psychiatric disorders for therapeutic purposes. Hence, ongoing clinical trials implicate synthetic analogs of pregnenolone or allopregnanolone, and also modulators of neurosteroidogenesis.
Collapse
Affiliation(s)
- Giovanni Tomaselli
- INSERM U1215, Neurocentre Magendie, Group "Physiopathology and Therapeutic Approaches of Stress-Related Disease", 146 Rue Léo Saignat, 33000 Bordeaux, France; University of Bordeaux, 33000 Bordeaux, France
| | - Monique Vallée
- INSERM U1215, Neurocentre Magendie, Group "Physiopathology and Therapeutic Approaches of Stress-Related Disease", 146 Rue Léo Saignat, 33000 Bordeaux, France; University of Bordeaux, 33000 Bordeaux, France.
| |
Collapse
|
24
|
Erol A, Ho AMC, Winham SJ, Karpyak VM. Sex hormones in alcohol consumption: a systematic review of evidence. Addict Biol 2019; 24:157-169. [PMID: 29280252 PMCID: PMC6585852 DOI: 10.1111/adb.12589] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/20/2017] [Accepted: 11/20/2017] [Indexed: 02/02/2023]
Abstract
Sex hormones play an important role in establishing sex‐distinctive brain structural and functional variations that could contribute to the sex differences in alcohol consumption behavior. Here, we systematically reviewed articles that studied sex hormone impacts on alcohol consumption and alcohol use disorder (AUD). An extensive literature search conducted in MEDLINE, PubMed, Scopus and CINAHL databases identified 776 articles, which were then evaluated for pre‐specified criteria for relevance and quality assurance. A total of 50 articles, including 19 human studies and 31 animal studies, were selected for this review. Existing evidence supports the association of increased testosterone level and increased risk for alcohol use and AUD in males but results are inconclusive in females. In contrast, the evidence supports the association of increased estrogen level and increased alcohol use in females, with mixed findings reported in males. Much less is known about the impact of progestins on alcohol use and misuse in human subjects. Future observational and experimental studies conducted in both sexes with a comprehensive hormone panel are needed to elucidate the impact of the interplay between various sex hormone levels during various developmental stages on alcohol use‐related phenotypes and AUD.
Collapse
Affiliation(s)
- Almila Erol
- Department of Psychiatry and Psychology; Mayo Clinic; Rochester MN USA
- Department of Psychiatry; Ataturk Education and Research Hospital; Turkey
| | - Ada M.-C. Ho
- Department of Psychiatry and Psychology; Mayo Clinic; Rochester MN USA
- Department of Molecular Pharmacology and Experimental Therapeutics; Mayo Clinic; Rochester MN USA
| | - Stacey J. Winham
- Department of Health Sciences Research; Mayo Clinic; Rochester MN USA
| | - Victor M. Karpyak
- Department of Psychiatry and Psychology; Mayo Clinic; Rochester MN USA
| |
Collapse
|
25
|
Milivojevic V, Covault J, Angarita GA, Siedlarz K, Sinha R. Neuroactive steroid levels and cocaine use chronicity in men and women with cocaine use disorder receiving progesterone or placebo. Am J Addict 2018; 28:16-21. [PMID: 30537098 DOI: 10.1111/ajad.12828] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/29/2018] [Accepted: 11/18/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Neuroactive steroids (NAS) may play a role in addiction, with observed increases in response to acute stress and drug use, but decreases with chronic substance use, suggesting that NAS neuroadaptations may occur with chronic substance use. However, levels of NAS in addicted individuals have not been systematically examined. Here, we evaluated a panel of NAS in men and women with cocaine use disorder (CUD) who participated in a clinical laboratory study of progesterone. METHODS Forty six CUD individuals were enrolled in a randomized placebo-controlled laboratory study to evaluate progesterone effects on levels of various NAS. On day 5 of a 7-day inpatient treatment regimen of 400 mg/day progesterone (15M/8F) or placebo (14M/9F), plasma levels of NAS known to be downstream of progesterone (allopregnanolone, pregnanolone), and NAS not in the progesterone synthesis pathway (androstanediol, testosterone, dehydroepiandrosterone [DHEA] and the NAS precursor, pregnenolone) were analyzed using highly sensitive gas chromatography/mass spectrometry (GC/MS). The relationship between each of the NAS and chronicity of cocaine use was also assessed. RESULTS Progesterone versus placebo significantly increased the GABAergic NAS allopregnanolone and pregnanolone in both CUD men and women. Levels of pregnenolone, testosterone, its GABAergic metabolite androstanediol, and the non-GABAergic DHEA were unaffected by progesterone treatment, and testosterone and androstanediol levels were significantly higher in men than women. Importantly, lower pregnenolone and androstanediol levels were associated with greater years of cocaine use. SCIENTIFIC SIGNIFICANCE GABAergic NAS that are upstream from the progesterone synthesis pathway appear susceptible to chronic effects of cocaine use. (Am J Addict 2019;28:16-21).
Collapse
Affiliation(s)
- Verica Milivojevic
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Jonathan Covault
- Alcohol Research Center, Department of Psychiatry, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Gustavo A Angarita
- Connecticut Mental Health Center, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Kristen Siedlarz
- Alcohol Research Center, Department of Psychiatry, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Rajita Sinha
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
26
|
Rice NC, Rauscher NA, Langston JL, Myers TM. Behavioral intoxication following voluntary oral ingestion of tetramethylenedisulfotetramine: Dose-dependent onset, severity, survival, and recovery. Neurotoxicology 2017; 63:21-32. [PMID: 28855111 DOI: 10.1016/j.neuro.2017.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 01/13/2023]
Abstract
Tetramethylenedisulfotetramine (tetramine, or TETS) is a highly toxic rodenticide that has been responsible for over 14,000 accidental and intentional poisonings worldwide. Although the vast majority of TETS poisonings involved tainted food or drink, the laboratory in vivo studies of TETS intoxication used intraperitoneal injection or gavage for TETS exposure. Seeking to develop and characterize a more realistic model of TETS intoxication in the present study, rats were trained to rapidly and voluntarily consume a poisoned food morsel. Initially, the overt toxic effects of TETS consumption across a large range of doses were characterized, then a focused range of doses was selected for more intensive behavioral evaluation (in operant test chambers providing a variable-interval schedule of food reinforcement). The onset of intoxication following voluntary oral consumption of TETS was rapid, and clear dose-dependent response-rate suppression was observed across multiple performance measures within the operant-chamber environment. At most doses, recovery of operant performance did not occur within 30h. Food consumption and body weight changes were also dose dependent and corroborated the behavioral measures of intoxication. This voluntary oral-poisoning method with concomitant operant-behavioral assessment shows promise for future studies of TETS (and other toxic chemicals of interest) and may be extremely valuable in characterizing treatment outcomes.
Collapse
Affiliation(s)
- Nathaniel C Rice
- United States Army Medical Research Institute of Chemical Defense, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD, 21010, USA
| | - Noah A Rauscher
- United States Army Medical Research Institute of Chemical Defense, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD, 21010, USA
| | - Jeffrey L Langston
- United States Army Medical Research Institute of Chemical Defense, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD, 21010, USA
| | - Todd M Myers
- United States Army Medical Research Institute of Chemical Defense, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD, 21010, USA.
| |
Collapse
|
27
|
Bell RL, Hauser SR, Liang T, Sari Y, Maldonado-Devincci A, Rodd ZA. Rat animal models for screening medications to treat alcohol use disorders. Neuropharmacology 2017; 122:201-243. [PMID: 28215999 PMCID: PMC5659204 DOI: 10.1016/j.neuropharm.2017.02.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 02/02/2017] [Accepted: 02/05/2017] [Indexed: 01/21/2023]
Abstract
The purpose of this review is to present animal research models that can be used to screen and/or repurpose medications for the treatment of alcohol abuse and dependence. The focus will be on rats and in particular selectively bred rats. Brief introductions discuss various aspects of the clinical picture, which provide characteristics of individuals with alcohol use disorders (AUDs) to model in animals. Following this, multiple selectively bred rat lines will be described and evaluated in the context of animal models used to screen medications to treat AUDs. Next, common behavioral tests for drug efficacy will be discussed particularly as they relate to stages in the addiction cycle. Tables highlighting studies that have tested the effects of compounds using the respective techniques are included. Wherever possible the Tables are organized chronologically in ascending order to describe changes in the focus of research on AUDs over time. In general, high ethanol-consuming selectively bred rats have been used to test a wide range of compounds. Older studies usually followed neurobiological findings in the selected lines that supported an association with a propensity for high ethanol intake. Most of these tests evaluated the compound's effects on the maintenance of ethanol drinking. Very few compounds have been tested during ethanol-seeking and/or relapse and fewer still have assessed their effects during the acquisition of AUDs. Overall, while a substantial number of neurotransmitter and neuromodulatory system targets have been assessed; the roles of sex- and age-of-animal, as well as the acquisition of AUDs, ethanol-seeking and relapse continue to be factors and behaviors needing further study. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Richard L Bell
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis, IN 46202, USA.
| | - Sheketha R Hauser
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis, IN 46202, USA
| | - Tiebing Liang
- Indiana University School of Medicine, Department of Gastroenterology, Indianapolis, IN 46202, USA
| | - Youssef Sari
- University of Toledo, Department of Pharmacology, Toledo, OH 43614, USA
| | | | - Zachary A Rodd
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis, IN 46202, USA
| |
Collapse
|
28
|
Beattie MC, Maldonado-Devincci AM, Porcu P, O'Buckley TK, Daunais JB, Grant KA, Morrow AL. Voluntary ethanol consumption reduces GABAergic neuroactive steroid (3α,5α)3-hydroxypregnan-20-one (3α,5α-THP) in the amygdala of the cynomolgus monkey. Addict Biol 2017; 22:318-330. [PMID: 26625954 DOI: 10.1111/adb.12326] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 08/31/2015] [Accepted: 10/04/2015] [Indexed: 01/27/2023]
Abstract
Neuroactive steroids such as (3α,5α)3-hydroxypregnan-20-one (3α,5α-THP, allopregnanolone) enhance the gamma-aminobutyric acid (GABA)-ergic effects of ethanol and modulate excessive drinking in rodents. Moreover, chronic ethanol consumption reduces 3α,5α-THP levels in human plasma, rat hippocampus and mouse limbic regions. We explored the relationship between 3α,5α-THP levels in limbic brain areas and voluntary ethanol consumption in the cynomolgus monkey following daily self-administration of ethanol for 12 months and further examined the relationship to hypothalamic-pituitary-adrenal (HPA) axis function prior to ethanol exposure. Monkeys were subjected to scheduled induction of ethanol consumption followed by free access to ethanol or water for 22 h/day over 12 months. Immunohistochemistry was performed using an anti-3α,5α-THP antibody. Prolonged voluntary drinking resulted in individual differences in ethanol consumption that ranged from 1.2 to 4.2 g/kg/day over 12 months. Prolonged ethanol consumption reduced cellular 3α,5α-THP immunoreactivity by 13 ± 2 percent (P < 0.05) in the lateral amygdala and 17 ± 2 percent (P < 0.05) in the basolateral amygdala. The effect of ethanol was most pronounced in heavy drinkers that consumed ≥3 g/kg ≥ 20 percent of days. Consequently, 3α,5α-THP immunoreactivity in both the lateral and basolateral amygdala was inversely correlated with average daily ethanol intake (Spearman r = -0.87 and -0.72, respectively, P < 0.05). However, no effect of ethanol and no correlation between drinking and 3α,5α-THP immunoreactivity were observed in the basomedial amygdala. 3α,5α-THP immunoreactivity following ethanol exposure was also correlated with HPA axis function prior to ethanol exposure. These data indicate that voluntary ethanol drinking reduces amygdala levels of 3α,5α-THP in non-human primates and that amygdala 3α,5α-THP levels may be linked to HPA axis function.
Collapse
Affiliation(s)
- Matthew C. Beattie
- Department of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies; University of North Carolina at Chapel Hill; Chapel Hill NC USA
| | - Antoniette M. Maldonado-Devincci
- Department of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies; University of North Carolina at Chapel Hill; Chapel Hill NC USA
| | - Patrizia Porcu
- Department of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies; University of North Carolina at Chapel Hill; Chapel Hill NC USA
- Neuroscience Institute; National Research Council of Italy (CNR); Italy
| | - Todd K. O'Buckley
- Department of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies; University of North Carolina at Chapel Hill; Chapel Hill NC USA
| | - James B. Daunais
- Department of Physiology and Pharmacology; Wake Forest School of Medicine; Winston Salem NC USA
| | - Kathleen A. Grant
- Division of Neuroscience, Oregon National Primate Research Center; Oregon Health and Science University; Beaverton OR USA
| | - A. Leslie Morrow
- Department of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies; University of North Carolina at Chapel Hill; Chapel Hill NC USA
| |
Collapse
|
29
|
Porcu P, O'Buckley TK, Lopez MF, Becker HC, Miles MF, Williams RW, Morrow AL. Initial genetic dissection of serum neuroactive steroids following chronic intermittent ethanol across BXD mouse strains. Alcohol 2017; 58:107-125. [PMID: 27884493 DOI: 10.1016/j.alcohol.2016.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 06/30/2016] [Accepted: 07/07/2016] [Indexed: 10/20/2022]
Abstract
Neuroactive steroids modulate alcohol's impact on brain function and behavior. Ethanol exposure alters neuroactive steroid levels in rats, humans, and some mouse strains. We conducted an exploratory analysis of the neuroactive steroids (3α,5α)-3-hydroxypregnan-20-one (3α,5α-THP), (3α,5α)-3,21-dihydroxypregnan-20-one (3α,5α-THDOC), and pregnenolone across 126-158 individuals and 19 fully inbred strains belonging to the BXD family, which were subjected to air exposure, or chronic intermittent ethanol (CIE) exposure. Neuroactive steroids were measured by gas chromatography-mass spectrometry in serum following five cycles of CIE or air exposure (CTL). Pregnenolone levels in CTLs range from 272 to 578 pg/mL (strain variation of 2.1 fold with p = 0.049 for strain main effect), with heritability of 0.20 ± 0.006 (SEM), whereas in CIE cases values range from 304 to 919 pg/mL (3.0-fold variation, p = 0.007), with heritability of 0.23 ± 0.005. 3α,5α-THP levels in CTLs range from 375 to 1055 pg/mL (2.8-fold variation, p = 0.0007), with heritability of 0.28 ± 0.01; in CIE cases they range from 460 to 1022 pg/mL (2.2-fold variation, p = 0.004), with heritability of 0.23 ± 0.005. 3α,5α-THDOC levels in CTLs range from 94 to 448 pg/mL (4.8-fold variation, p = 0.002), with heritability of 0.30 ± 0.01, whereas levels in CIE cases do not differ significantly. However, global averages across all BXD strains do not differ between CTL and CIE for any of the steroids. 3α,5α-THDOC levels were lower in females than males in both groups (CTL -53%, CIE -55%, p < 0.001). Suggestive quantitative trait loci are identified for pregnenolone and 3α,5α-THP levels. Genetic variation in 3α,5α-THP was not correlated with two-bottle choice ethanol consumption in CTL or CIE-exposed animals. However, individual variation in 3α,5α-THP correlated negatively with ethanol consumption in both groups. Moreover, strain variation in neuroactive steroid levels correlated with numerous behavioral phenotypes of anxiety sensitivity accessed in GeneNetwork, consistent with evidence that neuroactive steroids modulate anxiety-like behavior.
Collapse
|
30
|
Newman EL, Gunner G, Huynh P, Gachette D, Moss S, Smart T, Rudolph U, DeBold JF, Miczek KA. Effects of Gabra2 Point Mutations on Alcohol Intake: Increased Binge-Like and Blunted Chronic Drinking by Mice. Alcohol Clin Exp Res 2016; 40:2445-2455. [PMID: 27717041 PMCID: PMC5073020 DOI: 10.1111/acer.13215] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 08/11/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND Alcohol use disorders are associated with single-nucleotide polymorphisms in GABRA2, the gene encoding the GABAA receptor α2-subunit in humans. Deficient GABAergic functioning is linked to impulse control disorders, intermittent explosive disorder, and to drug abuse and dependence, yet it remains unclear whether α2-containing GABAA receptor sensitivity to endogenous ligands is involved in excessive alcohol drinking. METHODS Male wild-type (Wt) C57BL/6J and point-mutated mice rendered insensitive to GABAergic modulation by benzodiazepines (BZD; H101R), allopregnanolone (ALLO) or tetrahydrodeoxycorticosterone (THDOC; Q241M), or high concentrations of ethanol (EtOH) (S270H/L277A) at α2-containing GABAA receptors were assessed for their binge-like, moderate, or escalated chronic drinking using drinking in the dark, continuous access (CA) and intermittent access (IA) to alcohol protocols, respectively. Social approach by mutant and Wt mice in forced alcohol abstinence was compared to approach by EtOH-naïve controls. Social deficits in forced abstinence were treated with allopregnanolone (0, 3.0, 10.0 mg/kg, intraperitoneal [i.p.]) or midazolam (0, 0.56, 1.0 mg/kg, i.p.). RESULTS Mice with BZD-insensitive α2-containing GABAA receptors (H101R) escalated their binge-like drinking. Mutants harboring the Q241M point substitution in Gabra2 showed blunted chronic intake in the CA and IA protocols. S270H/L277A mutants consumed excessive amounts of alcohol but, unlike wild-types, they did not show forced abstinence-induced social deficits. CONCLUSIONS These findings suggest a role for: (i) H101 in species-typical binge-like drinking, (ii) Q241 in escalated chronic drinking, and (iii) S270 and/or L277 in the development of forced abstinence-associated social deficits. Clinical findings report reduced BZD-binding sites in the cortex of dependent patients; the present findings suggest a specific role for BZD-sensitive α2-containing receptors. In addition, amino acid residue 241 in Gabra2 is necessary for positive modulation and activation of GABAA receptors by ALLO and THDOC; we postulate that neurosteroid action on α2-containing receptor may be necessary for escalated chronic EtOH intake.
Collapse
Affiliation(s)
| | | | | | | | | | - Trevor Smart
- Dept. of Neuroscience, Physiology and Pharmacology, University College London
| | - Uwe Rudolph
- Laboratory of Genetic Neuropharmacology, McLean Hospital,Dept. of Psychiatry, Harvard Medical School
| | | | - Klaus A. Miczek
- Dept. of Psychology, Tufts University,Dept. of Neuroscience, Tufts University
| |
Collapse
|
31
|
Dury AY, Ke Y, Labrie F. Precise and accurate assay of pregnenolone and five other neurosteroids in monkey brain tissue by LC-MS/MS. Steroids 2016; 113:64-70. [PMID: 27378657 DOI: 10.1016/j.steroids.2016.06.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 06/28/2016] [Accepted: 06/30/2016] [Indexed: 11/20/2022]
Abstract
A series of steroids present in the brain have been named "neurosteroids" following the possibility of their role in the central nervous system impairments such as anxiety disorders, depression, premenstrual dysphoric disorder (PMDD), addiction, or even neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Study of their potential role requires a sensitive and accurate assay of their concentration in the monkey brain, the closest model to the human. We have thus developed a robust, precise and accurate liquid chromatography-tandem mass spectrometry method for the assay of pregnenolone, pregnanolone, epipregnanolone, allopregnanolone, epiallopregnanolone, and androsterone in the cynomolgus monkey brain. The extraction method includes a thorough sample cleanup using protein precipitation and phospholipid removal, followed by hexane liquid-liquid extraction and a Girard T ketone-specific derivatization. This method opens the possibility of investigating the potential implication of these six steroids in the most suitable animal model for neurosteroid-related research.
Collapse
Affiliation(s)
- Alain Y Dury
- Endoceutics Inc., 2795 Laurier Blvd, Suite 500, Quebec City (QC) G1V 4M7, Canada
| | - Yuyong Ke
- Endoceutics Inc., 2795 Laurier Blvd, Suite 500, Quebec City (QC) G1V 4M7, Canada
| | - Fernand Labrie
- Endoceutics Inc., 2795 Laurier Blvd, Suite 500, Quebec City (QC) G1V 4M7, Canada.
| |
Collapse
|
32
|
Spence AL, Guerin GF, Goeders NE. The differential effects of alprazolam and oxazepam on methamphetamine self-administration in rats. Drug Alcohol Depend 2016; 166:209-17. [PMID: 27485488 DOI: 10.1016/j.drugalcdep.2016.07.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 07/13/2016] [Accepted: 07/14/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Methamphetamine is the second most commonly used illicit drug in the world, and despite recent attempts by the Drug Enforcement Administration to combat this epidemic, methamphetamine use is still on the rise. As methamphetamine use increases so does polydrug use, particularly that involving methamphetamine and benzodiazepines. The present study was designed to examine the effects of two benzodiazepines on methamphetamine self-administration. METHODS Five doses of methamphetamine (0.0075, 0.015, 0.03, 0.09, and 0.12mg/kg/infusion) were tested, producing an inverted U-shaped dose-response curve. Rats were then pretreated with oxazepam, alprazolam, or vehicle prior to methamphetamine self-administration. To determine if the effects of these drugs were due to the GABAA receptor and/or translocator protein (TSPO), we also pretreated rats with an antagonist for the benzodiazepine-binding site on the GABAA receptor (i.e., flumazenil) and a TSPO antagonist (i.e., PK11195) prior to alprazolam or oxazepam administration. RESULTS Oxazepam significantly reduced methamphetamine self-administration as demonstrated by a downward shift of the dose-response curve. In contrast, alprazolam significantly enhanced methamphetamine self-administration as evidenced by a leftward shift of the dose-response curve. Flumazenil completely blocked the effects of alprazolam on methamphetamine self-administration. When administered individually, both flumazenil and PK11195 partially reversed the effects of oxazepam on methamphetamine self-administration. However, when these two antagonists were combined, the effects of oxazepam were completely reversed. CONCLUSIONS The GABAA receptor is responsible for the alprazolam-induced enhancement of methamphetamine self-administration, while the activation of both the GABAA receptor and TSPO are responsible for the oxazepam-induced reduction of methamphetamine self-administration.
Collapse
Affiliation(s)
- Allyson L Spence
- Department of Pharmacology, Toxicology, & Neuroscience, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA 71130, United States.
| | - Glenn F Guerin
- Department of Pharmacology, Toxicology, & Neuroscience, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA 71130, United States
| | - Nicholas E Goeders
- Department of Pharmacology, Toxicology, & Neuroscience, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA 71130, United States
| |
Collapse
|
33
|
Humble SR. Neurosteroids are reduced in diabetic neuropathy and may be associated with the development of neuropathic pain. F1000Res 2016; 5:1923. [PMID: 28357038 PMCID: PMC5345788 DOI: 10.12688/f1000research.9034.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/29/2016] [Indexed: 01/10/2023] Open
Abstract
Introduction: Peripheral and central sensitisation are implicated in the development of neuropathic pain. Hypersensitivity of pain pathway neurons has been described in animal models of diabetic neuropathy, which is postulated to be related to an imbalance between inhibitory and excitatory signals within the spinal cord. GABAergic neurons within the pain pathway are vital for the transmission of painful stimuli to higher centres. A developmental change in the rate of exponential decay of GABAergic synaptic events has been observed in other types of neurons and this may be associated with fluctuations in endogenous neurosteroid tone. Methods: The whole-cell patch-clamp technique was used on slices of neural tissue. Electrophysiological recordings were obtained from wild type mice between the ages of 6 and 80 days in the spinal cord, the nucleus reticularis of the thalamus and the cerebral cortex. Recordings were also obtained from mice with diabetic neuropathy (ob/ob and db/db) between the ages of 60 and 80 days. Behavioural experiments were performed to examine mechanical and thermal nociception. Results: Electrophysiological recordings from cortical pain pathway neurons from mature type-2 diabetic mice revealed that the endogenous neurosteroid tone is reduced compared to control. However, selected neurosteroid compounds had a more pronounced effect on the GABA
A receptors of these diabetic mice. ob/ob mice exhibit mechanical hyperalgesia and allodynia, which was reduced by neurosteroids applied exogenously. Conclusions: The reduced endogenous neurosteroid tone in ob/ob mice may be linked to their hypersensitivity. Neurosteroids may exert analgesic effects in pathological pain states by attempting to restore the physiological GABAergic inhibitory tone.
Collapse
Affiliation(s)
- Stephen R Humble
- Department of Anaesthetics and Pain Management, Charing Cross Hospital, Imperial College NHS Healthcare Trust London, London, W6 8RF, UK
| |
Collapse
|
34
|
Vallée M. Neurosteroids and potential therapeutics: Focus on pregnenolone. J Steroid Biochem Mol Biol 2016; 160:78-87. [PMID: 26433186 DOI: 10.1016/j.jsbmb.2015.09.030] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/18/2015] [Accepted: 09/21/2015] [Indexed: 12/26/2022]
Abstract
Considerable evidence from preclinical and clinical studies shows that steroids and in particular neurosteroids are important endogenous modulators of several brain-related functions. In this context, it remains to be elucidated whether neurosteroids may serve as biomarkers in the diagnosis of disorders and might have therapeutic potential for the treatment of these disorders. Pregnenolone (PREG) is the main steroid synthesized from cholesterol in mammals and invertebrates. PREG has three main sources of synthesis, the gonads, adrenal glands and brain and is submitted to various metabolizing pathways which are modulated depending on various factors including species, steroidogenic tissues and steroidogenic enzymes. Looking at the whole picture of steroids, PREG is often known as the precursor to other steroids and not as an active steroid per se. Actually, physiological and brain functions have been studied mainly for steroids that are very active either binding to specific intracellular receptors, or modulating with high affinity the abundant membrane receptors, GABAA or NMDA receptors. However, when high sensitive and specific methodological approaches were available to analyze low concentrations of steroids and then match endogenous levels of different steroid metabolomes, several studies have reported more significant alterations in PREG than in other steroids in extraphysiological or pathological conditions, suggesting that PREG could play a functional role as well. Additionally, several molecular targets of PREG were revealed in the mammalian brain and beneficial effects of PREG have been demonstrated in preclinical and clinical studies. On this basis, this review will be divided into three parts. The first provides a brief overview of the molecular targets of PREG and the pharmacological effects observed in animal and human studies. The second will focus on the possible functional role of PREG with an outline of the modulation of PREG levels in animal and in human research. Finally, the review will highlight the possible therapeutic uses of PREG that point towards the development of pregnenolone-like molecules.
Collapse
Affiliation(s)
- Monique Vallée
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux F33077, France; Université de Bordeaux, Bordeaux F33077, France.
| |
Collapse
|
35
|
Kärkkäinen O, Häkkinen MR, Auriola S, Kautiainen H, Tiihonen J, Storvik M. Increased steroid hormone dehydroepiandrosterone and pregnenolone levels in post-mortem brain samples of alcoholics. Alcohol 2016; 52:63-70. [PMID: 27139239 DOI: 10.1016/j.alcohol.2016.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 03/03/2016] [Accepted: 03/04/2016] [Indexed: 11/29/2022]
Abstract
Intra-tissue levels of steroid hormones (e.g., dehydroepiandrosterone [DHEA], pregnenolone [PREGN], and testosterone [T]) may influence the pathological changes seen in neurotransmitter systems of alcoholic brains. Our aim was to compare levels of these steroid hormones between the post-mortem brain samples of alcoholics and non-alcoholic controls. We studied steroid levels with quantitative liquid chromatography-tandem mass spectrometry (LC-MS/MS) in post-mortem brain samples of alcoholics (N = 14) and non-alcoholic controls (N = 10). Significant differences were observed between study groups in DHEA and PREGN levels (p values 0.0056 and 0.019, respectively), but not in T levels. Differences between the study groups were most prominent in the nucleus accumbens (NAC), anterior cingulate cortex (ACC), and anterior insula (AINS). DHEA levels were increased in most alcoholic subjects compared to controls. However, only a subgroup of alcoholics showed increased PREGN levels. Negative Spearman correlations between tissue levels of PREGN and previous reports of [(3)H]naloxone binding to μ-opioid receptors were observed in the AINS, ACC, NAC, and frontal cortex (R values between -0.6 and -0.8; p values ≤ 0.002), suggesting an association between the opioid system and brain PREGN levels. Although preliminary, and from relatively small diagnostic groups, these results show significantly increased levels of DHEA and PREGN in the brains of alcoholics, and could be associated with the pathology of alcoholism.
Collapse
Affiliation(s)
- Olli Kärkkäinen
- Pharmacology and Toxicology, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| | - Merja R Häkkinen
- Pharmaceutical Chemistry, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Seppo Auriola
- Pharmaceutical Chemistry, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Hannu Kautiainen
- General Practice, University of Helsinki, FI-00014 Helsinki, Finland; Unit of Primary Health Care, Kuopio University Hospital, FI-70029 Kuopio, Finland
| | - Jari Tiihonen
- Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, FI-70240 Kuopio, Finland; Clinical Neuroscience, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Markus Storvik
- Pharmacology and Toxicology, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| |
Collapse
|
36
|
Maldonado-Devincci AM, Kampov-Polevoi A, McKinley RE, Morrow DH, O'Buckley TK, Morrow AL. Chronic Intermittent Ethanol Exposure Alters Stress Effects on (3α,5α)-3-hydroxy-pregnan-20-one (3α,5α-THP) Immunolabeling of Amygdala Neurons in C57BL/6J Mice. Front Cell Neurosci 2016; 10:40. [PMID: 26973459 PMCID: PMC4777881 DOI: 10.3389/fncel.2016.00040] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/02/2016] [Indexed: 12/02/2022] Open
Abstract
The GABAergic neuroactive steroid (3α,5α)-3-hydroxy-pregnan-20-one (3α,5α-THP, allopregnanolone) is decreased in various brain regions of C57BL/6J mice following exposure to an acute stressor or chronic intermittent ethanol (CIE) exposure and withdrawal. It is well established that there are complex interactions between stress and ethanol drinking, with mixed literature regarding the effects of stress on ethanol intake. However, there is little research examining how chronic ethanol exposure alters stress responses. The present work examined the impact of CIE exposure and withdrawal on changes in brain levels of 3α,5α-THP, as well as hormonal and behavioral responses to forced swim stress (FSS). Adult male C57BL/6J mice were exposed to four cycles of CIE to induce ethanol dependence. Following 8 h or 72 h withdrawal, mice were subjected to FSS for 10 min, and 50 min later brains were collected for immunohistochemical analysis of cellular 3α,5α-THP. Behavioral and circulating corticosterone responses to FSS were quantified. Following 8 h withdrawal, ethanol exposure potentiated the corticosterone response to FSS. Following 72 h withdrawal, this difference was no longer observed. Following 8 h withdrawal, stress-exposed mice showed no differences in immobility, swimming or struggling behavior. However, following 72 h withdrawal, ethanol-exposed mice showed less immobility and greater swimming behavior compared to air-exposed mice. Interestingly, cellular 3α,5α-THP levels were increased in the lateral amygdala 8 h and 72 h post-withdrawal in stressed ethanol-exposed mice compared to ethanol-exposed/non-stressed mice. In the paraventricular nucleus of the hypothalamus, stress exposure decreased 3α,5α-THP levels compared to controls following 72 h withdrawal, but no differences were observed 8 h post-withdrawal. There were no differences in cellular 3α,5α-THP levels in the nucleus accumbens shell at either withdrawal time point. These data suggest that there are different mechanisms mediating hormonal, behavioral, and brain responses to stress following CIE exposure. The lateral amygdala appears to be an extremely sensitive brain region exhibiting changes in cellular 3α,5α-THP levels following CIE and exposure to swim stress. It is likely that these changes in cellular 3α,5α-THP levels in the lateral amygdala contribute to the behavioral effects observed following 72 h withdrawal.
Collapse
Affiliation(s)
| | - Alexander Kampov-Polevoi
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill Chapel Hill, NC, USA
| | - Raechel E McKinley
- Department of Biology, North Carolina Agricultural and Technical State University Greensboro, NC, USA
| | - Danielle H Morrow
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill Chapel Hill, NC, USA
| | - Todd K O'Buckley
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill Chapel Hill, NC, USA
| | - A Leslie Morrow
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel HillChapel Hill, NC, USA; Department of Psychiatry, University of North Carolina at Chapel HillChapel Hill, NC, USA; Department of Pharmacology, University of North Carolina at Chapel HillChapel Hill, NC, USA
| |
Collapse
|
37
|
Porcu P, Barron AM, Frye CA, Walf AA, Yang SY, He XY, Morrow AL, Panzica GC, Melcangi RC. Neurosteroidogenesis Today: Novel Targets for Neuroactive Steroid Synthesis and Action and Their Relevance for Translational Research. J Neuroendocrinol 2016; 28:12351. [PMID: 26681259 PMCID: PMC4769676 DOI: 10.1111/jne.12351] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 12/12/2015] [Accepted: 12/12/2015] [Indexed: 12/19/2022]
Abstract
Neuroactive steroids are endogenous neuromodulators synthesised in the brain that rapidly alter neuronal excitability by binding to membrane receptors, in addition to the regulation of gene expression via intracellular steroid receptors. Neuroactive steroids induce potent anxiolytic, antidepressant, anticonvulsant, sedative, analgesic and amnesic effects, mainly through interaction with the GABAA receptor. They also exert neuroprotective, neurotrophic and antiapoptotic effects in several animal models of neurodegenerative diseases. Neuroactive steroids regulate many physiological functions, such as the stress response, puberty, the ovarian cycle, pregnancy and reward. Their levels are altered in several neuropsychiatric and neurological diseases and both preclinical and clinical studies emphasise a therapeutic potential of neuroactive steroids for these diseases, whereby symptomatology ameliorates upon restoration of neuroactive steroid concentrations. However, direct administration of neuroactive steroids has several challenges, including pharmacokinetics, low bioavailability, addiction potential, safety and tolerability, which limit its therapeutic use. Therefore, modulation of neurosteroidogenesis to restore the altered endogenous neuroactive steroid tone may represent a better therapeutic approach. This review summarises recent approaches that target the neuroactive steroid biosynthetic pathway at different levels aiming to promote neurosteroidogenesis. These include modulation of neurosteroidogenesis through ligands of the translocator protein 18 kDa and the pregnane xenobiotic receptor, as well as targeting of specific neurosteroidogenic enzymes such as 17β-hydroxysteroid dehydrogenase type 10 or P450 side chain cleavage. Enhanced neurosteroidogenesis through these targets may be beneficial not only for neurodegenerative diseases, such as Alzheimer's disease and age-related dementia, but also for neuropsychiatric diseases, including alcohol use disorders.
Collapse
Affiliation(s)
- Patrizia Porcu
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Anna M. Barron
- Molecular Imaging Center, National Institute of Radiological Sciences, Anagawa, Inage-ku, Chiba, Japan
| | - Cheryl Anne Frye
- Institute of Arctic Biology, The University of Alaska–Fairbanks, Fairbanks, AK, USA
- The University at Albany, Albany, NY, USA
| | - Alicia A. Walf
- Institute of Arctic Biology, The University of Alaska–Fairbanks, Fairbanks, AK, USA
- The University at Albany, Albany, NY, USA
- Department of Cognitive Science, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Song-Yu Yang
- Department of Developmental Biochemistry, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Xue-Ying He
- Department of Developmental Biochemistry, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - A. Leslie Morrow
- Departments of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Gian Carlo Panzica
- Department of Neuroscience, University of Turin, and NICO - Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Roberto C. Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
38
|
Ford MM, Nickel JD, Kaufman MN, Finn DA. Null mutation of 5α-reductase type I gene alters ethanol consumption patterns in a sex-dependent manner. Behav Genet 2015; 45:341-53. [PMID: 25416204 PMCID: PMC4425631 DOI: 10.1007/s10519-014-9694-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 11/10/2014] [Indexed: 02/04/2023]
Abstract
The neuroactive steroid allopregnanolone (ALLO) is a positive modulator of GABAA receptors, and manipulation of neuroactive steroid levels via injection of ALLO or the 5α-reductase inhibitor finasteride alters ethanol self-administration patterns in male, but not female, mice. The Srd5a1 gene encodes the enzyme 5α-reductase-1, which is required for the synthesis of ALLO. The current studies investigated the influence of Srd5a1 deletion on voluntary ethanol consumption in male and female wildtype (WT) and knockout (KO) mice. Under a continuous access condition, 6 and 10 % ethanol intake was significantly greater in KO versus WT females, but significantly lower in KO versus WT males. In 2-h limited access sessions, Srd5a1 deletion retarded acquisition of 10 % ethanol intake in female mice, but facilitated it in males, versus respective WT mice. The present findings demonstrate that the Srd5a1 gene modulates ethanol consumption in a sex-dependent manner that is also contingent upon ethanol access condition and concentration.
Collapse
Affiliation(s)
- Matthew M Ford
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA,
| | | | | | | |
Collapse
|
39
|
Ramaker MJ, Strong-Kaufman MN, Ford MM, Phillips TJ, Finn DA. Effect of nucleus accumbens shell infusions of ganaxolone or gaboxadol on ethanol consumption in mice. Psychopharmacology (Berl) 2015; 232:1415-26. [PMID: 25342197 PMCID: PMC4412309 DOI: 10.1007/s00213-014-3777-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 10/10/2014] [Indexed: 01/16/2023]
Abstract
RATIONALE Allopregnanolone (ALLO) is an endogenous neuroactive steroid thought to alter the reinforcement value of alcohol (ethanol) due to its actions as a positive modulator of the GABAA receptor (GABAAR). Extrasynaptic GABAARs may be a particularly sensitive target of ethanol and neuroactive steroids. Previous work showed that systemic injections of an ALLO analog, ganaxolone (GAN), or an extrasynaptic GABAAR agonist (gaboxadol; THIP) decreased ethanol intake in male mice with limited access to ethanol. OBJECTIVES The present studies tested whether activation of GABAARs in the nucleus accumbens (NAc) shell by GAN or THIP was sufficient to reduce ethanol intake. C57BL/6J male mice had 2-h access to 10 % ethanol (10E) and water, and 10E intake was measured following site-specific infusions of GAN or THIP. RESULTS Decreases in limited-access 10E consumption were observed following site-specific bilateral infusions of either drug into the NAc shell. Significant changes in intake were absent when the drugs were infused in a region dorsal to the target site (GAN) or into the lateral ventricle (THIP). Locomotor data confirmed that the decreases in intake were not due to a sedative effect of the drugs. CONCLUSIONS These data demonstrate the sufficiency of GABAAR activation by a positive allosteric modulator or an agonist with selectivity for extrasynaptic GABAARs to decrease ethanol consumption in mice. Importantly, more refined GABAAR-active targets that decrease ethanol intake may enhance our understanding and ability to treat alcohol use disorders.
Collapse
Affiliation(s)
- Marcia J Ramaker
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road (L470), Portland, OR, 97239, USA,
| | | | | | | | | |
Collapse
|
40
|
Maldonado-Devincci AM, Cook JB, O'Buckley TK, Morrow DH, McKinley RE, Lopez MF, Becker HC, Morrow AL. Chronic intermittent ethanol exposure and withdrawal alters (3α,5α)-3-hydroxy-pregnan-20-one immunostaining in cortical and limbic brain regions of C57BL/6J mice. Alcohol Clin Exp Res 2014; 38:2561-71. [PMID: 25293837 DOI: 10.1111/acer.12530] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 07/15/2014] [Indexed: 02/03/2023]
Abstract
BACKGROUND The GABAergic neuroactive steroid (3α,5α)-3-hydroxy-pregnan-20-one (3α,5α-THP; allopregnanolone) has been studied during withdrawal from ethanol (EtOH) in humans, rats, and mice. Serum 3α,5α-THP levels decreased, and brain levels were not altered following acute EtOH administration (2 g/kg) in male C57BL/6J mice; however, the effects of chronic intermittent ethanol (CIE) exposure on 3α,5α-THP levels have not been examined. Given that CIE exposure changes subsequent voluntary EtOH drinking in a time-dependent fashion following repeated cycles of EtOH exposure, we conducted a time-course analysis of CIE effects on 3α,5α-THP levels in specific brain regions known to influence drinking behavior. METHODS Adult male C57BL/6J mice were exposed to 4 cycles of CIE to induce EtOH dependence. All mice were sacrificed and perfused at 1 of 2 time points, 8 or 72 hours following the final exposure cycle. Free-floating brain sections (40 μm; 3 to 5 sections/region/animal) were immunostained and analyzed to determine relative levels of cellular 3α,5α-THP. RESULTS Withdrawal from CIE exposure produced time-dependent and region-specific effects on immunohistochemical detection of 3α,5α-THP levels across cortical and limbic brain regions. A transient reduction in 3α,5α-THP immunoreactivity was observed in the central nucleus of the amygdala 8 hours after withdrawal from CIE (-31.4 ± 9.3%). Decreases in 3α,5α-THP immunoreactivity were observed 72 hours following withdrawal in the medial prefrontal cortex (-25.0 ± 9.3%), nucleus accumbens core (-29.9 ± 6.6%), and dorsolateral striatum (-18.5 ± 6.0%), while an increase was observed in the CA3 pyramidal cell layer of the hippocampus (+42.8 ± 19.5%). Sustained reductions in 3α,5α-THP immunoreactivity were observed at both time points in the lateral amygdala (8 hours -28.3 ± 12.8%; 72 hours -27.5 ± 12.4%) and in the ventral tegmental area (8 hours -26.5 ± 9.9%; 72 hours -31.6 ± 13.8%). CONCLUSIONS These data suggest that specific neuroadaptations in 3α,5α-THP levels may be present in regions of brain that mediate anxiety, stress, and reinforcement relevant to EtOH dependence. The changes that occur at different time points likely modulate neurocircuitry involved in EtOH withdrawal as well as the elevated drinking observed after CIE exposure.
Collapse
Affiliation(s)
- Antoniette M Maldonado-Devincci
- Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Effects of the neuroactive steroid allopregnanolone on intracranial self-stimulation in C57BL/6J mice. Psychopharmacology (Berl) 2014; 231:3415-3423. [PMID: 24810108 PMCID: PMC4692244 DOI: 10.1007/s00213-014-3600-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 04/20/2014] [Indexed: 10/25/2022]
Abstract
RATIONALE The neuroactive steroid (3α,5α)-3-hydroxy-pregnan-20-one (3α,5α-THP, allopregnanolone) has effects on reward-related behaviors in mice and rats that suggest that it may activate brain reward circuits. Intracranial self-stimulation (ICSS) is an operant behavioral technique that detects changes in the sensitivity of brain reward circuitry following drug administration. OBJECTIVE To examine the effects of the neuroactive steroid allopregnanolone on ICSS and to compare these effects to those of cocaine. METHODS Male C57BL/6J mice implanted with stimulating electrodes implanted into the medial forebrain bundle responded for reinforcement by electrical stimulation (brain stimulation reward (BSR)). Mice received cocaine (n = 11, 3.0-30.0 mg/kg, intraperitoneal (i.p.)) or the neuroactive steroid allopregnanolone (n = 11, 3.0-17.0 mg/kg, i.p.). BSR thresholds (θ 0) and maximum (MAX) operant response rates after drug treatments were compared to those after vehicle injections. RESULTS Cocaine and allopregnanolone dose dependently lowered BSR thresholds relative to vehicle injections. Cocaine was maximally effective (80 % reduction) in the second 15 min following the 30 mg/kg dose, while allopregnanolone was maximally effective (30 % reduction) 15-45 min after the 17 mg/kg dose. Neither drug had significant effects on MAX response rates. CONCLUSIONS The effects of allopregnanolone on BSR thresholds are consistent with the previously reported effects of benzodiazepines and alcohol, suggesting that positive modulation of GABAA receptors can facilitate reward-related behaviors in C57BL/6J mice.
Collapse
|
42
|
Porcu P, Morrow AL. Divergent neuroactive steroid responses to stress and ethanol in rat and mouse strains: relevance for human studies. Psychopharmacology (Berl) 2014; 231:3257-72. [PMID: 24770626 PMCID: PMC4135033 DOI: 10.1007/s00213-014-3564-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 03/25/2014] [Indexed: 01/09/2023]
Abstract
RATIONALE Neuroactive steroids are endogenous or synthetic steroids that rapidly alter neuronal excitability via membrane receptors, primarily γ-aminobutyric acid type A (GABAA) receptors. Neuroactive steroids regulate many physiological processes including hypothalamic-pituitary-adrenal (HPA) axis function, ovarian cycle, pregnancy, aging, and reward. Moreover, alterations in neuroactive steroid synthesis are implicated in several neuropsychiatric disorders. OBJECTIVES This review will summarize the pharmacological properties and physiological regulation of neuroactive steroids, with a particular focus on divergent neuroactive steroid responses to stress and ethanol in rats, mice, and humans. RESULTS GABAergic neuroactive steroids exert a homeostatic regulation of the HPA axis in rats and humans, whereby the increase in neuroactive steroid levels following acute stress counteracts HPA axis hyperactivity and restores homeostasis. In contrast, in C57BL/6J mice, acute stress decreases neurosteroidogenesis and neuroactive steroids exert paradoxical excitatory effects upon the HPA axis. Rats, mice, and humans also differ in the neuroactive steroid responses to ethanol. Genetic variation in neurosteroidogenesis may explain the different neuroactive steroid responses to stress or ethanol. CONCLUSIONS Rats and mouse strains show divergent effects of stress and ethanol on neuroactive steroids in both plasma and brain. The study of genetic variation in the various processes that determine neuroactive steroids levels as well as their effects on cell signaling may underlie these differences and may play a relevant role for the potential therapeutic benefits of neuroactive steroids.
Collapse
Affiliation(s)
- Patrizia Porcu
- Neuroscience Institute, National Research Council of Italy (CNR), Cittadella Universitaria, 09042, Monserrato, Cagliari, Italy,
| | | |
Collapse
|
43
|
Ramaker MJ, Ford MM, Phillips TJ, Finn DA. Differences in the reinstatement of ethanol seeking with ganaxolone and gaboxadol. Neuroscience 2014; 272:180-7. [PMID: 24814021 PMCID: PMC4122668 DOI: 10.1016/j.neuroscience.2014.04.065] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 04/21/2014] [Accepted: 04/24/2014] [Indexed: 10/25/2022]
Abstract
The endogenous neuroactive steroid allopregnanolone (ALLO) has previously been shown to induce reinstatement of ethanol seeking in rodents. ALLO is a positive allosteric modulator at both synaptic and extrasynaptic GABAA receptors. The contribution of each class of GABAA receptors in mediating reinstatement of ethanol seeking is unknown. The first aim of the present study was to determine whether ganaxolone (GAN), a longer-acting synthetic analog of ALLO, also promotes reinstatement of ethanol seeking. The second aim was to examine whether preferentially activating extrasynaptic GABAA receptors with the selective agonist gaboxadol (THIP) was sufficient to reinstate responding for ethanol in mice. Male C57BL/6J mice were trained to lever press for access to a 10% ethanol (v/v) solution (10E), using a sucrose-fading procedure. Following extinction of the lever-pressing behavior, systemic THIP (0, 4 and 6mg/kg) and GAN (0, 10, and 15mg/kg) were tested for their ability to reinstate ethanol-appropriate responding in the absence of 10E access. GAN significantly increased lever pressing on the previously active lever, while THIP did not alter lever-pressing behavior. The results of this study suggest that direct activation of extrasynaptic GABAA receptors at the GABA site is not sufficient to induce ethanol seeking in the reinstatement procedure. Future studies are necessary to elucidate the mechanisms and brain areas by which differences in the pharmacological activity of GAN and THIP at the GABAA receptor contribute to the dissimilarity in their effect on the reinstatement of ethanol seeking. Nonetheless, based on the increased use of these drugs in clinical trials across multiple disease states, the effects of GAN or THIP on alcohol seeking may be an important consideration if these drugs are to be used clinically in a population with a co-occurring alcohol use disorder.
Collapse
Affiliation(s)
- M J Ramaker
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States.
| | - M M Ford
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States
| | - T J Phillips
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States; Department of Veterans Affairs Medical Research, Portland, OR 97239, United States
| | - D A Finn
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States; Department of Veterans Affairs Medical Research, Portland, OR 97239, United States
| |
Collapse
|
44
|
Rezvani AH, Levin ED. Assessment of pregnenolone effects on alcohol intake and preference in male alcohol preferring (P) rats. Eur J Pharmacol 2014; 740:53-7. [PMID: 25016089 DOI: 10.1016/j.ejphar.2014.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 06/30/2014] [Accepted: 07/01/2014] [Indexed: 01/16/2023]
Abstract
Neuroactive steroids can modulate a variety of neurobehavioral functions via the GABAergic system. This study was conducted to determine the importance of the neurosteroid pregnenolone on the regulation of alcohol intake. The effects of acute and chronic administration of pregnenolone on alcohol intake were assessed in alcohol preferring (P) rats. The rats were injected i.p. with the vehicle or pregnenolone (25, 50 or 75 mg/kg) and their alcohol and water intake were recorded at 2, 4, 6 and 24 h. Also, the chronic effects of 50 mg/kg (i.p.) pregnenolone on alcohol intake were determined. Our results show that although the main effect of i.p. injection of pregnenolone in reducing alcohol intake was not quite significant compared with the vehicle, pregnenolone at 75 mg/kg significantly (P<0.025) reduced alcohol intake. Regarding alcohol preference, acute administration of pregnenolone both at 50 mg/kg (P<0.05) and at 75 mg/kg (P<0.025) significantly reduced alcohol preference. In chronic experiments pregnenolone given for 10 consecutive days did not show a significant effect on alcohol intake and alcohol preference. Overall, although pregnenolone given i.p. acutely and significantly reduced alcohol intake and preference, the fact that chronic treatment did not show an effect diminishes its promise to be considered for the treatment of alcoholism. However, its profile of effects might be different in human alcoholics.
Collapse
Affiliation(s)
- Amir H Rezvani
- Department of Psychiatry and Behavioral Sciences and Department of Psychology and Neuroscience, Duke University Medical Center, Durham, NC 27710, USA.
| | - Edward D Levin
- Department of Psychiatry and Behavioral Sciences and Department of Psychology and Neuroscience, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
45
|
Overexpression of the steroidogenic enzyme cytochrome P450 side chain cleavage in the ventral tegmental area increases 3α,5α-THP and reduces long-term operant ethanol self-administration. J Neurosci 2014; 34:5824-34. [PMID: 24760842 DOI: 10.1523/jneurosci.4733-13.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neuroactive steroids are endogenous neuromodulators capable of altering neuronal activity and behavior. In rodents, systemic administration of endogenous or synthetic neuroactive steroids reduces ethanol self-administration. We hypothesized this effect arises from actions within mesolimbic brain regions that we targeted by viral gene delivery. Cytochrome P450 side chain cleavage (P450scc) converts cholesterol to pregnenolone, the rate-limiting enzymatic reaction in neurosteroidogenesis. Therefore, we constructed a recombinant adeno-associated serotype 2 viral vector (rAAV2), which drives P450scc expression and neuroactive steroid synthesis. The P450scc-expressing vector (rAAV2-P450scc) or control GFP-expressing vector (rAAV2-GFP) were injected bilaterally into the ventral tegmental area (VTA) or nucleus accumbens (NAc) of alcohol preferring (P) rats trained to self-administer ethanol. P450scc overexpression in the VTA significantly reduced ethanol self-administration by 20% over the 3 week test period. P450scc overexpression in the NAc, however, did not alter ethanol self-administration. Locomotor activity was unaltered by vector administration to either region. P450scc overexpression produced a 36% increase in (3α,5α)-3-hydroxypregnan-20-one (3α,5α-THP, allopregnanolone)-positive cells in the VTA, but did not increase 3α,5α-THP immunoreactivity in NAc. These results suggest that P450scc overexpression and the resultant increase of 3α,5α-THP-positive cells in the VTA reduces ethanol reinforcement. 3α,5α-THP is localized to neurons in the VTA, including tyrosine hydroxylase neurons, but not astrocytes. Overall, the results demonstrate that using gene delivery to modulate neuroactive steroids shows promise for examining the neuronal mechanisms of moderate ethanol drinking, which could be extended to other behavioral paradigms and neuropsychiatric pathology.
Collapse
|
46
|
Anton RF, Schacht JP, Book SW. Pharmacologic treatment of alcoholism. HANDBOOK OF CLINICAL NEUROLOGY 2014; 125:527-42. [PMID: 25307594 DOI: 10.1016/b978-0-444-62619-6.00030-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Progress in understanding the neuroscience of addiction has significantly advanced the development of more efficacious medications for the treatment of alcohol use disorders (AUD). While several medications have been approved by regulatory bodies around the world for the treatment of AUD, they are not universally efficacious. Recent research has yielded improved understanding of the genetics and brain circuits that underlie alcohol reward and its habitual use. This research has contributed to pharmacogenetic studies of medication response, and will ultimately lead to a more "personalized medicine" approach to AUD pharmacotherapy. This chapter summarizes work on clinically available medications (both approved by regulatory bodies and investigational) for the treatment of alcohol dependence, as well as the psychiatric disorders that are commonly comorbid with AUD. Studies that have evaluated genetic influences on medication response and those that have employed neuroimaging to probe mechanisms of medication action or response are highlighted. Finally, new targets discovered in animal models for possible pharmacologic intervention in humans are overviewed and future directions in medications development provided.
Collapse
Affiliation(s)
- Raymond F Anton
- Center for Drug and Alcohol Programs, Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA.
| | - Joseph P Schacht
- Center for Drug and Alcohol Programs, Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Sarah W Book
- Center for Drug and Alcohol Programs, Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
47
|
Cook JB, Dumitru AMG, O'Buckley TK, Morrow AL. Ethanol administration produces divergent changes in GABAergic neuroactive steroid immunohistochemistry in the rat brain. Alcohol Clin Exp Res 2013; 38:90-9. [PMID: 23906006 DOI: 10.1111/acer.12223] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 05/06/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND The 5α-reduced pregnane neuroactive steroid (3α,5α)-3-hydroxypregnan-20-one (3α,5α-THP or allopregnanolone) is a potent positive modulator of GABAA receptors capable of modulating neuronal activity. In rats, systemic ethanol (EtOH) administration increases cerebral cortical and hippocampal levels of 3α,5α-THP, but the effects of EtOH on 3α,5α-THP levels in other brain regions are unknown. There is a large body of evidence suggesting that 3α,5α-THP enhances EtOH sensitivity, contributes to some behavioral effects of EtOH, and modulates EtOH reinforcement and motivation to drink. In this study, we used immunohistochemistry (IHC) to determine EtOH-induced changes in cellular 3α,5α-THP expression in brain regions associated with EtOH actions and responses. METHODS Male Wistar rats were administered EtOH (2 g/kg) or saline intraperitoneally and after 60 minutes transcardially perfused. IHC was performed on free-floating sections (3 to 4 sections/animal/brain region) using an affinity purified anti-3α,5α-THP primary antibody, and immunoreactivity was visualized with 3,3'-diaminobenzidine. RESULTS EtOH significantly increased 3α,5α-THP immunoreactivity by 24 ± 6% in the medial prefrontal cortex, 32 ± 12% in the hippocampal Cornu Ammonis area 1 (CA1) pyramidal cell layer, 52 ± 5% in the polymorph cell layer of the dentate gyrus (DG), 44 ± 15% in the bed nucleus of the stria terminalis, and 36 ± 6% in the paraventricular nucleus of the hypothalamus. In contrast, EtOH administration significantly reduced 3α,5α-THP immunoreactivity by 25 ± 5% in the nucleus accumbens "shore" and 21 ± 3% in the central nucleus of the amygdala. No changes were observed in the ventral tegmental area, dorsomedial striatum, granule cell layer of the DG, or the lateral and basolateral amygdala. CONCLUSIONS The results suggest acute EtOH (2 g/kg) produces divergent, brain region specific, effects on cellular 3α,5α-THP levels. Regional differences in the effects of EtOH suggest there may be regional brain synthesis of 3α,5α-THP independent of the adrenal glands and novel mechanisms that reduce cellular 3α,5α-THP. Regional differences in EtOH-induced changes in 3α,5α-THP levels likely contribute to EtOH effects on neuronal function in brain.
Collapse
Affiliation(s)
- Jason B Cook
- Departments of Psychiatry and Pharmacology , Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | | |
Collapse
|
48
|
Milivojevic V, Covault J. Alcohol exposure during late adolescence increases drinking in adult Wistar rats, an effect that is not reduced by finasteride. Alcohol Alcohol 2013; 48:28-38. [PMID: 22997410 PMCID: PMC3523383 DOI: 10.1093/alcalc/ags105] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 06/18/2012] [Accepted: 07/31/2012] [Indexed: 11/13/2022] Open
Abstract
AIMS We tested whether an exposure to alcohol in late adolescence, an age of rapid increase in neuroactive steroid precursors, would increase voluntary alcohol consumption in adult rats and whether this effect would be modulated by finasteride, an inhibitor of neuroactive steroid synthesis. METHODS In Experiment 1, we exposed male Wistar rats to 8% alcohol during the dark cycle for 1 week during late adolescence [postnatal days (PNDs) 51-58], and then measured voluntary alcohol consumption 1 month later in adulthood (PNDs 91-104). In Experiment 2, finasteride was administered during the forced alcohol exposure in late adolescence and, in Experiment 3, during voluntary alcohol consumption in adulthood. Plasma was collected at the end of each finasteride treatment to confirm the reduction of plasma neuroactive steroid levels. RESULTS We found that a daily 12-h exposure to alcohol for 7 days in late adolescence significantly increased voluntary alcohol consumption (4-fold) a month later during adulthood. Finasteride administration in late adolescence increased group alcohol intake in late adolescence but did not block the effect of adolescent alcohol exposure on increasing alcohol preference in adulthood. There was no effect of finasteride treatment in adulthood on alcohol preference. CONCLUSIONS A daily 12-h exposure to alcohol for 7 days in late adolescence was sufficient to induce chronically increased alcohol preference in adulthood, indicating that this age may be sensitive to the effects of alcohol.
Collapse
Affiliation(s)
- Verica Milivojevic
- Graduate Program in Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Psychiatry, Alcohol Research Center, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Jonathan Covault
- Department of Psychiatry, Alcohol Research Center, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
49
|
Davies DL, Bortolato M, Finn DA, Ramaker MJ, Barak S, Ron D, Liang J, Olsen RW. Recent advances in the discovery and preclinical testing of novel compounds for the prevention and/or treatment of alcohol use disorders. Alcohol Clin Exp Res 2012; 37:8-15. [PMID: 22671690 DOI: 10.1111/j.1530-0277.2012.01846.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 03/26/2012] [Indexed: 11/27/2022]
Abstract
Alcohol abuse and dependence have a staggering socioeconomic impact, yet current therapeutic strategies are largely inadequate to treat these disorders. Thus, the development of new strategies that can effectively prevent alcohol use disorders (AUDs) is of paramount importance. Currently approved medications attempt to deter alcohol intake by blocking ethanol metabolism or by targeting the neurochemical systems downstream of the cascades leading to craving and dependence. Unfortunately, these medications have provided only limited success as indicated by the continued high rates of alcohol abuse and alcoholism. The lack of currently available effective treatment strategies is highlighted by the urgent call by the NIAAA to find new and paradigm-changing therapeutics to either prevent or treat alcohol-related problems. This mini-review highlights recent findings from 4 laboratories with a focus on compounds that have the potential to be novel therapeutic agents that can be developed for the prevention and/or treatment of AUDs.
Collapse
Affiliation(s)
- Daryl L Davies
- School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Ramaker MJ, Strong MN, Ford MM, Finn DA. Effect of ganaxolone and THIP on operant and limited-access ethanol self-administration. Neuropharmacology 2012; 63:555-64. [PMID: 22613838 DOI: 10.1016/j.neuropharm.2012.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 05/04/2012] [Accepted: 05/06/2012] [Indexed: 11/18/2022]
Abstract
Recent evidence suggests that GABA(A) receptor ligands may regulate ethanol intake via effects at both synaptic and extrasynaptic receptors. For example, the endogenous neurosteroid, allopregnanolone (ALLO) has a similar pharmacological profile as ethanol, and it alters ethanol intake in rodent models. Additionally, recent evidence suggests that δ-subunit-containing extrasynaptic GABA(A) receptors may confer high sensitivity to both ethanol and neurosteroids. The purpose of the present study was to determine the effects of ganaxolone (GAN; an ALLO analog) and gaboxadol (THIP; a GABA(A) receptor agonist with selectivity for the extrasynaptic δ-subunit) on ethanol intake, drinking patterns, and bout characteristics in operant and limited-access self-administration procedures. In separate studies, the effects of GAN (0-10 mg/kg) and THIP (2-16 mg/kg) were tested in C57BL/6J male mice provided with 2-h access to a two-bottle choice of water or 10% ethanol or trained to respond for 30 min of access to 10% ethanol. GAN had no overall significant effect on operant ethanol self-administration, but tended to decrease the latency to consume the first bout. In the limited-access procedure, GAN dose-dependently decreased ethanol intake. THIP dose-dependently decreased ethanol intake in both paradigms, altering both the consummatory and appetitive processes of operant self-administration as well as shifting the drinking patterns in both procedures. These results add to literature suggesting time-dependent effects of neurosteroids to promote the onset, and to subsequently decrease, ethanol drinking behavior, and they support a role for extrasynaptic GABA(A) receptor activation in ethanol reinforcement.
Collapse
Affiliation(s)
- Marcia J Ramaker
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA.
| | | | | | | |
Collapse
|