1
|
Perovic M, Pavlovic D, Palmer Z, Udo MSB, Citadin CT, Rodgers KM, Wu CYC, Zhang Q, Lin HW, Tesic V. Modulation of GABAergic system as a therapeutic option in stroke. Exp Neurol 2025; 384:115050. [PMID: 39522803 DOI: 10.1016/j.expneurol.2024.115050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/18/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Stroke is one of the leading causes of death and permanent adult disability worldwide. Despite the improvements in reducing the rate and mortality, the societal burden and costs of treatment associated with stroke management are increasing. Most of the therapeutic approaches directly targeting ischemic injury have failed to reduce short- and long-term morbidity and mortality and more effective therapeutic strategies are still needed to promote post-stroke functional recovery. Decades of stroke research have been focused on hyperexcitability and glutamate-induced excitotoxicity in the acute phase of ischemia and their relation to motor deficits. Recent advances in understanding the pathophysiology of stroke have been made with several lines of evidence suggesting that changes in the neurotransmission of the major inhibitory system via γ-Aminobutyric acid (GABA) play a particularly important role in functional recovery and deserve further attention. The present review provides an overview of how GABAergic neurotransmission changes correlate with stroke recovery and outlines GABAergic system modulators with special emphasis on neurosteroids that have been shown to affect stroke pathogenesis or plasticity or to protect against cognitive decline. Supporting evidence from both animal and human clinical studies is presented and the potential for GABA signaling-targeted therapies for stroke is discussed to translate this concept to human neural repair therapies. Age and sex are considered crucial parameters related to the pathophysiology of stroke and important factors in the development of therapeutic pharmacological strategies. Future work is needed to deepen our knowledge of the neurochemical changes after stroke, extend the conceptual framework, and allow for the development of more effective interventions that include the modulation of the inhibitory system.
Collapse
Affiliation(s)
- Milka Perovic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Damjan Pavlovic
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Zoe Palmer
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Mariana S B Udo
- Department of Neurology, University of Texas Houston Health Science Center, TX, USA
| | - Cristiane T Citadin
- Department of Neurology, University of Texas Houston Health Science Center, TX, USA
| | - Krista M Rodgers
- Department of Cellular Biology and Anatomy, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Celeste Yin-Chien Wu
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Hung Wen Lin
- Department of Neurology, University of Texas Houston Health Science Center, TX, USA
| | - Vesna Tesic
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA.
| |
Collapse
|
2
|
Reshma A, Subramanian A, Kumarasamy V, Tamilanban T, Sekar M, Gan S, Subramaniyan V, Wong L, Rani N, Wu Y. Neurocognitive effects of proanthocyanidin in Alzheimer's disease: a systematic review of preclinical evidence. Braz J Med Biol Res 2024; 57:e13587. [PMID: 39504064 PMCID: PMC11540257 DOI: 10.1590/1414-431x2024e13587] [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: 12/04/2023] [Accepted: 09/17/2024] [Indexed: 11/08/2024] Open
Abstract
Cognitive disorders and dementia largely influence individual independence and orientation. Based on the Alzheimer's Disease International (ADI) estimation, approximately 75% of individuals with dementia are undiagnosed. In fact, in some low- and middle-income countries, the percentage is as high as 90%. In this systematic review, which is based on PRISMA guidelines, we aim to identify the mechanism of action of proanthocyanidin. Finding a natural product alternative as a potential nootropic can help increase the number of armamentariums against dementia and other cognitive impairments. In this preclinical research, we determined the effect of proanthocyanidins on Alzheimer's disease (AD) by searching electronic bibliographic databases like Scopus, Proquest, ScienceDirect, PubMed, and Google. There was no imposed time limit. However, the search was limited to only English articles. The review protocol is registered on PROSPERO as CRD42022356301. A population, intervention, control, and outcomes (PICO) technique was utilized for report inclusion, and all reports were assessed for risk of bias by using the SYRCLE's RoB tool. The article's bibliographic information, induction model, type of proanthocyanidins, animal strain/weight/age, and outcome measurements were acquired from ten papers and are reported here. Further analysis was validated and determined for the review. The included studies met the review's inclusion criteria and suggested that proanthocyanidins have a neurocognitive effect against AD. Additionally, the effectiveness of proanthocyanidins in reducing oxidative stress, acetylcholinesterase activity, amyloid beta, its efficacy in alleviating superoxide dismutase, cognitive properties, and in facilitating cholinergic transmission in various models of AD has been collectively observed in ten studies.
Collapse
Affiliation(s)
- A. Reshma
- Department of Pharmacology, SRM College of Pharmacy, SRM
Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamilnadu,
India
| | - A. Subramanian
- Department of Pharmacology, SRM College of Pharmacy, SRM
Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamilnadu,
India
| | - V. Kumarasamy
- Department of Parasitology & Medical Entomology, Faculty of
Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Kuala Lumpur,
Malaysia
| | - T. Tamilanban
- Department of Pharmacology, SRM College of Pharmacy, SRM
Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamilnadu,
India
- Department of Occupational Safety and Health, Faculty of Public
Health, Universitas Airlangga, Surabaya, Indonesia
- Faculty of Health and Life Sciences, INTI International
University, Nilai, Malaysia
- Department of Pharmacology, Faculty of Medicine, MAHSA
University, Bandar Saujana Putra, Selangor, Malaysia
| | - M. Sekar
- School of Pharmacy, Monash University Malaysia, Bandar Sunway,
Selangor, Malaysia
| | - S.H. Gan
- School of Pharmacy, Monash University Malaysia, Bandar Sunway,
Selangor, Malaysia
| | - V. Subramaniyan
- Department of Medical Sciences, School of Medical and Life
Sciences, Sunway University, Bandar Sunway, Selangor, Malaysia
| | - L.S. Wong
- Faculty of Health and Life Sciences, INTI International
University, Nilai, Malaysia
| | - N.N.I.M. Rani
- Faculty of Pharmacy and Health Sciences, Royal College of
Medicine Perak, Universiti Kuala Lumpur, Perak, Malaysia
| | - Y.S. Wu
- Sunway Microbiome Centre & Department of Biological
Sciences, School of Medical and Life Sciences, Sunway University, Subang Jaya,
Selangor, Malaysia
| |
Collapse
|
3
|
Fernandes E Mendonça LM, Joshi AB, Bhandarkar A, Shaikh S, Fernandes S, Joshi H, Joshi S. Potential anxiolytic therapeutics from Hybanthus enneaspermus (L.) F. Muell. - mitigate anxiety by plausibly modulating the GABA A-Cl - channel. Neurochem Int 2024; 178:105804. [PMID: 39002759 DOI: 10.1016/j.neuint.2024.105804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/13/2024] [Accepted: 07/09/2024] [Indexed: 07/15/2024]
Abstract
Anxiety is a commonly prevailing psychological disorder that requires effective treatment, wherein phytopharmaceuticals and nutraceuticals could offer a desirable therapeutic profile. Hybanthus enneaspermus (L.) F. Muell. is a powerful medicinal herb, reportedly effective against several ailments, including psychological disorders. The current research envisaged evaluating the anxiolytic potential of the ethanolic extract of Hybanthus enneaspermus (EEHE) and its toluene insoluble biofraction (ITHE) employing experimental and computational approaches. Elevated Plus Maze, Light and Dark Transition, Mirror Chamber, Hole board and Open field tests were used as screening models to assess the antianxiety potential of 100, 200 and 400 mg/kg body weight of EEHE and ITHE in rats subjected to social isolation, using Diazepam as standard. The brains of rats exhibiting significant anxiolytic activity were dissected for histopathological and biochemical studies. Antioxidant enzymes like catalase, superoxide dismutase, glutathione-S-transferase, glutathione peroxidase, glutathione reductase; and neurotransmitters viz. monoamines (serotonin, noradrenaline, dopamine), Gamma-aminobutyric acid (GABA), and glutamate were quantified in the different regions of rats' brain (cortex, hippocampus, pons, medulla oblongata, cerebellum). Chromatographic techniques were used to isolate phytoconstituents from the fraction exhibiting significant activity that were characterized by spectroscopic methods and subjected to in silico molecular docking. ITHE at 400 mg/kg body weight significantly mitigated anxiety in all the screening models (p < 0.05), reduced the inflammatory vacuoles and necrosis (p < 0.05) and potentiated the antioxidant enzymes (p < 0.05). It enhanced the monoamines and GABA levels while attenuating glutamate levels (p < 0.01) in the brain. Three significant flavonoids viz. Quercitrin, Rutin and Hesperidin were isolated from ITHE. In silico docking studies of these flavonoids revealed that the compounds exhibited substantial binding to the GABAA receptor. ITHE displayed a promising pharmacological profile in combating anxiety and modulating oxidative stress, attributing its therapeutic virtues to the flavonoids present.
Collapse
Affiliation(s)
| | - Arun Bhimrao Joshi
- Department of Pharmacognosy and Phytochemistry, Goa College of Pharmacy, 18th June Road, Panaji, Goa, 403001, India.
| | - Anant Bhandarkar
- Department of Pharmacognosy and Phytochemistry, Goa College of Pharmacy, 18th June Road, Panaji, Goa, 403001, India.
| | - Shamshad Shaikh
- School of Biological Sciences and Biotechnology, Goa University, Taleigao Plateau, Taleigao, Goa, 403206, India.
| | - Samantha Fernandes
- School of Biological Sciences and Biotechnology, Goa University, Taleigao Plateau, Taleigao, Goa, 403206, India.
| | - Himanshu Joshi
- Department of Pharmacology, College of Pharmacy, Graphic Era Hill University, Bhimtal Campus, Uttarakhand, 263156, India.
| | - Shrinivas Joshi
- Department of Pharmaceutical Chemistry, S.E.T.'s College of Pharmacy, Sangolli Rayanna Nagar, Dharwad, Karnataka, 580002, India.
| |
Collapse
|
4
|
Reddy DS, Mbilinyi RH, Estes E. Preclinical and clinical pharmacology of brexanolone (allopregnanolone) for postpartum depression: a landmark journey from concept to clinic in neurosteroid replacement therapy. Psychopharmacology (Berl) 2023; 240:1841-1863. [PMID: 37566239 PMCID: PMC10471722 DOI: 10.1007/s00213-023-06427-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/17/2023] [Indexed: 08/12/2023]
Abstract
This article describes the critical role of neurosteroids in postpartum depression (PPD) and outlines the landmark pharmacological journey of brexanolone as a first-in-class neurosteroid antidepressant with significant advantages over traditional antidepressants. PPD is a neuroendocrine disorder that affects about 20% of mothers after childbirth and is characterized by symptoms including persistent sadness, fatigue, dysphoria, as well as disturbances in cognition, emotion, appetite, and sleep. The main pathology behind PPD is the postpartum reduction of neurosteroids, referred to as neurosteroid withdrawal, a concept pioneered by our preclinical studies. We developed neurosteroid replacement therapy (NRT) as a rational approach for treating PPD and other conditions related to neurosteroid deficiency, unveiling the power of neurosteroids as novel anxiolytic-antidepressants. The neurosteroid, brexanolone (BX), is a progesterone-derived allopregnanolone that rapidly relieves anxiety and mood deficits by activating GABA-A receptors, making it a transformational treatment for PPD. In 2019, the FDA approved BX, an intravenous formulation of allopregnanolone, as an NRT to treat PPD. In clinical studies, BX significantly improved PPD symptoms within hours of administration, with tolerable side effects including headache, dizziness, and somnolence. We identified the molecular mechanism of BX in a neuronal PPD-like milieu. The mechanism of BX involves activation of both synaptic and extrasynaptic GABA-A receptors, which promote tonic inhibition and serve as a key target for PPD and related conditions. Neurosteroids offer several advantages over traditional antidepressants, including rapid onset, unique mechanism, and lack of tolerance upon repeated use. Some limitations of BX therapy include lack of aqueous solubility, limited accessibility, hospitalization for treatment, lack of oral product, and serious adverse events at high doses. However, the unmet need for synthetic neurosteroids to address this critical condition supersedes these limitations. Recently, we developed novel hydrophilic neurosteroids with a superior profile and improved drug delivery. Overall, approval of BX is a major milestone in the field of neurotherapeutics, paving the way for the development of novel synthetic neurosteroids to treat depression, epilepsy, and status epilepticus.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, 77807, USA.
- Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, 8447 Riverside Pkwy, Bryan, TX, 77807, USA.
| | - Robert H Mbilinyi
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, 77807, USA
| | - Emily Estes
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, 77807, USA
| |
Collapse
|
5
|
Saaiman D, Brand L, de Brouwe G, Janse van Rensburg H, Terre'Blanche G, Legoabe L, Krahe T, Wolmarans D. Striatal adenosine A 2A receptor involvement in normal and large nest building deer mice: perspectives on compulsivity and anxiety. Behav Brain Res 2023; 449:114492. [PMID: 37172739 DOI: 10.1016/j.bbr.2023.114492] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/25/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Obsessive-compulsive disorder (OCD) is characterized by recurring obsessive thoughts and repetitive behaviors that are often associated with anxiety and perturbations in cortico-striatal signaling. Given the suboptimal response of OCD to current serotonergic interventions, there is a need to better understand the psychobiological mechanisms that may underlie the disorder. In this regard, investigations into adenosinergic processes might be fruitful. Indeed, adenosine modulates both anxiety- and motor behavioral output. Thus, we aimed to explore the potential associations between compulsive-like large nest building (LNB) behavior in deer mice, anxiety and adenosinergic processes. From an initial pool of 120 adult deer mice, 34 normal nest building (NNB)- and 32 LNB-expressing mice of both sexes were selected and exposed to either a normal water (wCTRL) or vehicle control (vCTRL), lorazepam (LOR) or istradefylline (ISTRA) for 7- (LOR) or 28 days after which nesting assessment was repeated and animals screened for anxiety-like behavior in an anxiogenic open field. Mice were then euthanized, the striatal tissue removed on ice and the adenosine A2A receptor expression quantified. Our findings indicate that NNB and LNB behavior are not distinctly associated with measures of generalized anxiety and that ISTRA-induced changes in nesting expression are dissociated from changes in anxiety scores. Further, data from this investigation show that nesting in deer mice is directly related to striatal adenosine signaling, and that LNB is founded upon a lower degree of adenosinergic A2A stimulation.
Collapse
Affiliation(s)
- D Saaiman
- Center of Excellence for Pharmaceutical Sciences, Department of Pharmacology, North-West University, Potchefstroom, South Africa
| | - L Brand
- Center of Excellence for Pharmaceutical Sciences, Department of Pharmacology, North-West University, Potchefstroom, South Africa
| | - G de Brouwe
- Center of Excellence for Pharmaceutical Sciences, Department of Pharmacology, North-West University, Potchefstroom, South Africa
| | - H Janse van Rensburg
- Center of Excellence for Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, North-West University, Potchefstroom, South Africa
| | - G Terre'Blanche
- Center of Excellence for Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, North-West University, Potchefstroom, South Africa
| | - L Legoabe
- Center of Excellence for Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, North-West University, Potchefstroom, South Africa
| | - T Krahe
- Department of Psychology, Pontifical Catholic University of Rio de Janeiro (PUC-Rio), Rio de Janeiro, Brazil
| | - D Wolmarans
- Center of Excellence for Pharmaceutical Sciences, Department of Pharmacology, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
6
|
Caldiroli A, Capuzzi E, Affaticati LM, Surace T, Di Forti CL, Dakanalis A, Clerici M, Buoli M. Candidate Biological Markers for Social Anxiety Disorder: A Systematic Review. Int J Mol Sci 2023; 24:835. [PMID: 36614278 PMCID: PMC9821596 DOI: 10.3390/ijms24010835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/24/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023] Open
Abstract
Social anxiety disorder (SAD) is a common psychiatric condition associated with a high risk of psychiatric comorbidity and impaired social/occupational functioning when not promptly treated. The identification of biological markers may facilitate the diagnostic process, leading to an early and proper treatment. Our aim was to systematically review the available literature about potential biomarkers for SAD. A search in the main online repositories (PubMed, ISI Web of Knowledge, PsychInfo, etc.) was performed. Of the 662 records screened, 61 were included. Results concerning cortisol, neuropeptides and inflammatory/immunological/neurotrophic markers remain inconsistent. Preliminary evidence emerged about the role of chromosome 16 and the endomannosidase gene, as well as of epigenetic factors, in increasing vulnerability to SAD. Neuroimaging findings revealed an altered connectivity of different cerebral areas in SAD patients and amygdala activation under social threat. Some parameters such as salivary alpha amylase levels, changes in antioxidant defenses, increased gaze avoidance and QT dispersion seem to be associated with SAD and may represent promising biomarkers of this condition. However, the preliminary positive correlations have been poorly replicated. Further studies on larger samples and investigating the same biomarkers are needed to identify more specific biological markers for SAD.
Collapse
Affiliation(s)
- Alice Caldiroli
- Department of Mental Health and Addiction, Fondazione IRCCS San Gerardo dei Tintori, Via G.B. Pergolesi 33, 20900 Monza, Italy; (E.C.); (T.S.); (M.C.)
| | - Enrico Capuzzi
- Department of Mental Health and Addiction, Fondazione IRCCS San Gerardo dei Tintori, Via G.B. Pergolesi 33, 20900 Monza, Italy; (E.C.); (T.S.); (M.C.)
| | - Letizia M. Affaticati
- Department of Medicine and Surgery, University of Milano Bicocca, Via Cadore 38, 20900 Monza, Italy; (L.M.A.); (C.L.D.F.); (A.D.)
| | - Teresa Surace
- Department of Mental Health and Addiction, Fondazione IRCCS San Gerardo dei Tintori, Via G.B. Pergolesi 33, 20900 Monza, Italy; (E.C.); (T.S.); (M.C.)
| | - Carla L. Di Forti
- Department of Medicine and Surgery, University of Milano Bicocca, Via Cadore 38, 20900 Monza, Italy; (L.M.A.); (C.L.D.F.); (A.D.)
| | - Antonios Dakanalis
- Department of Medicine and Surgery, University of Milano Bicocca, Via Cadore 38, 20900 Monza, Italy; (L.M.A.); (C.L.D.F.); (A.D.)
| | - Massimo Clerici
- Department of Mental Health and Addiction, Fondazione IRCCS San Gerardo dei Tintori, Via G.B. Pergolesi 33, 20900 Monza, Italy; (E.C.); (T.S.); (M.C.)
- Department of Medicine and Surgery, University of Milano Bicocca, Via Cadore 38, 20900 Monza, Italy; (L.M.A.); (C.L.D.F.); (A.D.)
| | - Massimiliano Buoli
- Department of Pathophysiology and Transplantation, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy;
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milan, Italy
| |
Collapse
|
7
|
Watanabe S. Are mirrors aversive or rewarding for mice? Insights from the mirror preference test. Front Behav Neurosci 2023; 17:1137206. [PMID: 37122492 PMCID: PMC10133477 DOI: 10.3389/fnbeh.2023.1137206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
|
8
|
Wang YM, Xia CY, Jia HM, He J, Lian WW, Yan Y, Wang WP, Zhang WK, Xu JK. Sigma-1 receptor: A potential target for the development of antidepressants. Neurochem Int 2022; 159:105390. [PMID: 35810915 DOI: 10.1016/j.neuint.2022.105390] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/10/2022] [Accepted: 07/05/2022] [Indexed: 10/17/2022]
Abstract
Though a great many of studies on the development of antidepressants for the therapy of major depression disorder (MDD) and the development of antidepressants have been carried out, there still lacks an efficient approach in clinical practice. The involvement of Sigma-1 receptor in the pathological process of MDD has been verified. In this review, recent research focusing on the role of Sigma-1 receptor in the etiology of MDD were summarized. Preclinical studies and clinical trials have found that stress induce the variation of Sigma-1 receptor in the blood, brain and heart. Dysfunction and absence of Sigma-1 receptor result in depressive-like behaviors in rodent animals. Agonists of Sigma-1 receptor show not only antidepressant-like activities but also therapeutical effects in complications of depression. The mechanisms underlying antidepressant-like effects of Sigma-1 receptor may include suppressing neuroinflammation, regulating neurotransmitters, ameliorating brain-derived neurotrophic factor and N-Methyl-D-Aspartate receptor, and alleviating the endoplasmic reticulum stress and mitochondria damage during stress. Therefore, Sigma-1 receptor represents a potential target for antidepressants development.
Collapse
Affiliation(s)
- Yu-Ming Wang
- School of Life Sciences & School of Chinese Medicine Sciences, Beijing University of Chinese Medicine, Beijing, 100029, PR China; Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Cong-Yuan Xia
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Hong-Mei Jia
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, PR China
| | - Jun He
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Wen-Wen Lian
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Yu Yan
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Wen-Ping Wang
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Wei-Ku Zhang
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China.
| | - Jie-Kun Xu
- School of Life Sciences & School of Chinese Medicine Sciences, Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| |
Collapse
|
9
|
Ogaly HA, Abdel-Rahman RF, Mohamed MAE, O A AF, Khattab MS, Abd-Elsalam RM. Thymol ameliorated neurotoxicity and cognitive deterioration in a thioacetamide-induced hepatic encephalopathy rat model; involvement of the BDNF/CREB signaling pathway. Food Funct 2022; 13:6180-6194. [PMID: 35583008 DOI: 10.1039/d1fo04292k] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the present study, we aimed to delineate the neuroprotective potential of thymol (THY) against neurotoxicity and cognitive deterioration induced by thioacetamide (TAA) in an experimental model of hepatic encephalopathy (HE). Rats received TAA (100 mg kg-1, intraperitoneally injected, three times per week) for two weeks. THY (30 and 60 mg kg-1), and Vit E (100 mg k-1) were administered daily by oral gavage for 30 days after HE induction. Supplementation with THY significantly improved liver function, reduced serum ammonia level, and ameliorated the locomotor and cognitive deficits. THY effectively modulated the alteration in oxidative stress markers, neurotransmitters, and brain ATP content. Histopathology of liver and brain tissues showed that THY had ameliorated TAA-induced damage, astrocyte swelling and brain edema. Furthermore, THY downregulated NF-kB and upregulated GFAP protein expression. In addition, THY significantly promoted CREB and BDNF expression at both mRNA and protein levels, together with enhancing brain cAMP level. In conclusion, THY exerted hepato- and neuroprotective effects against HE by mitigating hepatotoxicity, hyperammonemia and brain ATP depletion via its antioxidant, anti-inflammatory effects in addition to activation of the CREB/BDNF signaling pathway.
Collapse
Affiliation(s)
- Hanan A Ogaly
- Department of Chemistry, College of Science, King Khalid University, Abha, Saudi Arabia. .,Department of Biochemistry, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Rehab F Abdel-Rahman
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Center, Dokki, Giza, Egypt
| | - Marawan Abd Elbaset Mohamed
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Center, Dokki, Giza, Egypt
| | - Ahmed-Farid O A
- Department of Physiology, National Organization for Drug Control and Research, Giza, Egypt
| | - Marwa S Khattab
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Reham M Abd-Elsalam
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
10
|
Abstract
BACKGROUND Anxiety disorders are highly prevalent affecting up to 33.7% of people over a lifetime. Although many treatment options are available, they are often associated with unacceptable side-effect profiles and approximately one in three patients are treatment resistant. Allopregnanolone, a neuroactive steroid acting as a positive allosteric modulator at the GABAA receptor, is synthesised in response to stress and acts to negatively modulate the hypothalamic-pituitary-adrenal axis. FINDINGS After chronic exposure to and withdrawal from allopregnanolone, an increase in α4β2δ GABAA receptors results in a reduced inhibitory effect of allopregnanolone, resulting in decreased inhibition and, therefore, increased neuronal excitability. The relationship between allopregnanolone and increased α4β2δ GABAA receptors has been demonstrated in animal models during methamphetamine withdrawal and puberty, events both associated with stress. The effect of allopregnanolone during these events is anxiogenic, a paradoxical action to its usual anxiolytic effects. Flumazenil, the GABAA receptor antagonist, has been shown to cause receptor internalisation of α4β2δ GABAA receptors, which may results in anxiolysis. CONCLUSION We propose that chronic stress and chronic exposure to and withdrawal from allopregnanolone in anxiety disorders result in alterations in GABAA receptor function, which can be corrected by flumazenil. As such, flumazenil may exhibit anxiolytic properties in patients with increased α4β2δ GABAA receptor expression.
Collapse
Affiliation(s)
- Alexander T Gallo
- Division of Psychiatry, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Gary K Hulse
- Division of Psychiatry, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Fresh Start Recovery Programme, Subiaco, WA, Australia
| |
Collapse
|
11
|
Reddy DS. Neurosteroid replacement therapy for catamenial epilepsy, postpartum depression and neuroendocrine disorders in women. J Neuroendocrinol 2022; 34:e13028. [PMID: 34506047 PMCID: PMC9247111 DOI: 10.1111/jne.13028] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/27/2021] [Accepted: 08/05/2021] [Indexed: 12/11/2022]
Abstract
Neurosteroids are involved in the pathophysiology of many neuroendocrine disorders in women. This review describes recent advancements in pharmacology of neurosteroids and emphasizes the benefits of neurosteroid replacement therapy for the management of neuroendocrine disorders such as catamenial epilepsy (CE), postpartum depression (PPD) and premenstrual brain conditions. Neurosteroids are endogenous modulators of neuronal excitability. A variety of neurosteroids are present in the brain including allopregnanolone (AP), allotetrahydro-deoxycorticosterone and androstanediol. Neurosteroids interact with synaptic and extrasynaptic GABAA receptors in the brain. AP and related neurosteroids, which are positive allosteric modulators of GABAA receptors, are powerful anticonvulsants, anxiolytic, antistress and neuroprotectant agents. In CE, seizures are most often clustered around a specific menstrual period in women. Neurosteroid withdrawal-linked plasticity in extrasynaptic receptors has been shown to play a key role in catamenial seizures, anxiety and other mood disorders. Based on our extensive research spanning two decades, we have proposed and championed neurosteroid replacement therapy as a rational strategy for treating disorders marked by neurosteroid-deficiency, such as CE and other related ovarian or menstrual disorders. In 2019, AP (renamed as brexanolone) was approved for treating PPD. A variety of synthetic neurosteroids are in clinical trials for epilepsy, depression and other brain disorders. Recent advancements in our understanding of neurosteroids have entered a new era of drug discovery and one that offers a high therapeutic potential for treating complex brain disorders.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, USA
| |
Collapse
|
12
|
Wolmarans DW, Prinsloo M, Seedat S, Stein DJ, Harvey BH, de Brouwer G. Escitalopram and lorazepam differentially affect nesting and open field behaviour in deer mice exposed to an anxiogenic environment. Neurosci Res 2021; 177:85-93. [PMID: 34736961 DOI: 10.1016/j.neures.2021.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/21/2021] [Accepted: 10/28/2021] [Indexed: 11/29/2022]
Abstract
Large nest building behaviour (LNB), as expressed by a subpopulation of laboratory housed deer mice (Peromyscus maniculatus bairdii), is persistent and repetitive. However, the response of LNB to an anxiogenic environment has not yet been investigated. Here, we employed LNB and normal nesting (NNB) expressing mice, subdivided into three drug-exposed groups per cohort, i.e. water (28 days), escitalopram (50 mg/kg/day, 28 days) and lorazepam (2 mg/kg/day; 4 days) to investigate this theme. During the last 4 days of drug exposure, mice were placed inside anxiogenic open field arenas which contained a separate enclosed and dark area for 4 consecutive nights during which open field and/or nest building assessments were performed. We show that LNB behaviour in deer mice is stable, irrespective of the anxiety-related context in which it is assessed, and that LNB mice find an open field arena to be less aversive compared to NNB mice. Escitalopram and lorazepam differentially affected the nesting and open field behaviour of LNB expressing mice, confirming deer mouse LNB as a repetitive behavioural phenotype that is related to a compulsive-like process which is regulated by the serotonergic system.
Collapse
Affiliation(s)
- De Wet Wolmarans
- Center of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West-University, Potchefstroom, South Africa.
| | - Michelle Prinsloo
- Center of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West-University, Potchefstroom, South Africa
| | - Soraya Seedat
- Department of Psychiatry, Stellenbosch University, Tygerberg, South Africa
| | - Dan J Stein
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, South Africa; MRC Unit on Risk and Resilience in Mental Disorders, Cape Town, South Africa
| | - Brian H Harvey
- Center of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West-University, Potchefstroom, South Africa; MRC Unit on Risk and Resilience in Mental Disorders, Cape Town, South Africa
| | - Geoffrey de Brouwer
- Center of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West-University, Potchefstroom, South Africa
| |
Collapse
|
13
|
Karim N, Khan I, Abdelhalim A, Halim SA, Khan A, Al-Harrasi A. Stigmasterol can be new steroidal drug for neurological disorders: Evidence of the GABAergic mechanism via receptor modulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 90:153646. [PMID: 34280827 DOI: 10.1016/j.phymed.2021.153646] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/22/2021] [Accepted: 06/26/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Gamma-aminobutyric acid A (GABAA) receptors have been implicated in anxiety and epileptic disorders. HYPOTHESIS/PURPOSE This study aimed to investigate the effects of stigmasterol, a plant sterol (phytosterol) isolated from Artemisia indica Linn on neurological disorders. METHODS Stigmasterol was evaluated on various recombinant GABAA receptor subtypes expressed in Xenopus laevis oocytes and its anxiolytic and anticonvulsant potential was assessed using the elevated plus maze (EPM), light-dark box (LDB) test, and pentylenetetrazole- (PTZ-) induced seizure paradigms. Furthermore, computational modeling of α2β2γ2L, α4β3δ, and α4β3 subtypes was performed to gain insights into the GABAergic mechanism of stigmasterol. For the first time, a model of GABAδ subtype was generated. Stigmasterol was targeted to all the binding sites (neurotransmitters, positive and negative modulator binding sites) of GABAA α2β2γ2L, α4β3, and α4β3δ complexes by in silico docking. RESULTS Stigmasterol enhanced GABA-induced currents at ternary α2β2γ2L, α4β3δ, and binary α4β3 GABAAR subtypes. The potentiation of GABA-induced currents at extrasynaptic α4β3δ was significantly higher compared to the binary α4β3 subtype, indicating that the δ subunit is important for efficacy. Stigmasterol was found to be a potent positive modulator of the extrasynaptic α4β3δ subtype, which was also confirmed by computational analysis. The computational analysis reveals that stigmasterol preferentially binds at the transmembrane region shared by positive modulators or a binding site constituted by the M2-M3 region of α4 and M1-M2 of β3 at α4β3δ complex. In in vivo studies, Stigmasterol (0.5-3.0 mg/kg, i.p.) exerted significant anxiolytic and anticonvulsant effects in an identical manner of allopregnanolone, indicating the involvement of a GABAergic mechanism. CONCLUSION To our knowledge, this is the first study reporting the positive modulation of GABAA receptors, anxiolytic and anticonvulsant potential of stigmasterol. Thus, stigmasterol is considered to be a candidate steroidal drug for the treatment of neurological disorders due to its positive modulation of GABA receptors.
Collapse
Affiliation(s)
- Nasiara Karim
- Department of Pharmacy, University of Malakand, Chakdara, Dir (Lower), KPK, Pakistan.
| | - Imran Khan
- Department of Pharmacy, University of Swabi, KPK, Pakistan
| | - Abeer Abdelhalim
- Faculty of Science, Taibah University, Almadina Almonawara, Saudi Arabia
| | - Sobia Ahsan Halim
- Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box 33, Birkat Al Mauz 616, Nizwa, Sultanate of Oman
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box 33, Birkat Al Mauz 616, Nizwa, Sultanate of Oman.
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, P.O. Box 33, Birkat Al Mauz 616, Nizwa, Sultanate of Oman.
| |
Collapse
|
14
|
Chen S, Gao L, Li X, Ye Y. Allopregnanolone in mood disorders: Mechanism and therapeutic development. Pharmacol Res 2021; 169:105682. [PMID: 34019980 DOI: 10.1016/j.phrs.2021.105682] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 01/23/2023]
Abstract
The neuroactive steroid allopregnanolone (ALLO) is an endogenous positive allosteric modulator of GABA type A receptor (GABAAR), and the down-regulation of its biosynthesis have been attributed to the development of mood disorders, such as depression, anxiety and post-traumatic stress disorder (PTSD). ALLO mediated depression/anxiety involves GABAergic mechanisms and appears to be related to brain-derived neurotrophic factor (BDNF), dopamine receptor, glutamate neurotransmission, and Ca2+ channel. In the clinical, brexanolone, as a newly developed intravenous ALLO preparation, has been approved for the treatment of postpartum depression (PPD). In addition, traditional antidepressants such as selective serotonin reuptake inhibitor (SSRI) could reverse ALLO decline. Recently, the translocation protein (TSPO, 18 kDa), which involves in the speed-limiting step of ALLO synthesis, and ALLO derivatization have been identified as new directions for antidepressant therapy. This review provides an overview of ALLO researches in animal model and patients, discusses its role in the development and treatment of depression/anxiety, and directs its therapeutic potential in future.
Collapse
Affiliation(s)
- Shiyi Chen
- School of Pharmacy, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, People's Republic of China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China.
| | - Lijuan Gao
- School of Pharmacy, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, People's Republic of China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China.
| | - Xiaoyu Li
- School of Pharmacy, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, People's Republic of China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China.
| | - Yiping Ye
- School of Pharmacy, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, People's Republic of China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
15
|
Frye CA, Qrareya A, Llaneza DC, Paris JJ. Central Actions of 3α,5α-THP Involving NMDA and GABA A Receptors Regulate Affective and Sexual Behavior of Female Rats. Front Behav Neurosci 2020; 14:11. [PMID: 32116591 PMCID: PMC7026732 DOI: 10.3389/fnbeh.2020.00011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 01/20/2020] [Indexed: 11/16/2022] Open
Abstract
The neurosteroid, 5α-pregnan-3α-ol-20-one (known as “allopregnanolone” or 3α,5α-THP), is produced in the midbrain ventral tegmental area (VTA), independent of peripheral sources of progestogens, where it has potential actions at N-methyl-D-aspartate (NMDA) and GABAA receptors to facilitate rodent sexual behavior. Progestogens can also have anti-anxiety effects, but whether these involve actions of centrally-derived 3α,5α-THP or these receptors to support reproductively-relevant behavior is not well understood. We investigated the extent to which 3α,5α-THP’s actions via NMDA and/or GABAA receptors in the midbrain VTA influence reproductive behaviors. Estradiol-primed, ovariectomized/adrenalectomized (OVX/ADX) rats received midbrain VTA infusions of vehicle, an NMDA receptor blocker (MK-801; 200 ng), or a GABAA receptor blocker (bicuculline; 100 ng) followed by a second infusion of vehicle or 3α,5α-THP (100 ng). Reproductively-relevant behaviors were assessed: sexual (paced mating), anxiety-like (elevated plus maze), and social (partner preference, social interaction) behavior. Compared to vehicle, intra-VTA infusions of MK-801 exerted anxiolytic-like effects on elevated plus maze behavior and enhanced lordosis. Unlike prior observations in gonadally-intact rats, intra-VTA bicuculline had no effect on the behavior of OVX/ADX rats (likely due to a floor effect). Subsequent infusions of 3α,5α-THP reversed effects on lordosis and infusions of bicuculline inhibited 3α,5α-THP-facilitated lordosis. Thus, NMDA and GABAA receptors may act as mediators for reproductive behavioral effects of 3α,5α-THP in the midbrain VTA.
Collapse
Affiliation(s)
- Cheryl A Frye
- Department of Psychology, The University at Albany-The State University of New York (SUNY), Albany, NY, United States.,Biological Sciences, The University at Albany-The State University of New York (SUNY), Albany, NY, United States.,Centers for Neuroscience, The University at Albany-The State University of New York (SUNY), Albany, NY, United States.,Life Sciences Research, The University at Albany-The State University of New York (SUNY), Albany, NY, United States
| | - Alaa Qrareya
- Department of Biomolecular Sciences, The University of Mississippi, University, MS, United States
| | - Danielle C Llaneza
- Department of Psychology, The University at Albany-The State University of New York (SUNY), Albany, NY, United States
| | - Jason J Paris
- Department of Psychology, The University at Albany-The State University of New York (SUNY), Albany, NY, United States.,Department of Biomolecular Sciences, The University of Mississippi, University, MS, United States
| |
Collapse
|
16
|
Samardzic J, Hencic B, Jancic J, Jadzic D, Djuric M, Obradovic DI, Svob Strac D. Neurosteroid dehydroepiandrosterone improves active avoidance retrieval and induces antidepressant-like behavior in rats. Neurosci Lett 2017; 660:17-21. [DOI: 10.1016/j.neulet.2017.09.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/01/2017] [Accepted: 09/07/2017] [Indexed: 10/18/2022]
|
17
|
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
|
18
|
Bartlett AA, Singh R, Hunter RG. Anxiety and Epigenetics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 978:145-166. [PMID: 28523545 DOI: 10.1007/978-3-319-53889-1_8] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Anxiety disorders are highly prevalent psychiatric disorders often comorbid with depression and substance abuse. Twin studies have shown that anxiety disorders are moderately heritable. Yet, genome-wide association studies (GWASs) have failed to identify gene(s) significantly associated with diagnosis suggesting a strong role for environmental factors and the epigenome. A number of anxiety disorder subtypes are considered "stress related." A large focus of research has been on the epigenetic and anxiety-like behavioral consequences of stress. Animal models of anxiety-related disorders have provided strong evidence for the role of stress on the epigenetic control of the hypothalamic-pituitary-adrenal (HPA) axis and of stress-responsive brain regions. Neuroepigenetics may continue to explain individual variation in susceptibility to environmental perturbations and consequently anxious behavior. Behavioral and pharmacological interventions aimed at targeting epigenetic marks associated with anxiety may prove fruitful in developing treatments.
Collapse
Affiliation(s)
- Andrew A Bartlett
- Department of Psychology, University of Massachusetts, 100 Morrissey Blvd, Boston, MA, 02125, USA
| | - Rumani Singh
- Department of Psychology, University of Massachusetts, 100 Morrissey Blvd, Boston, MA, 02125, USA
| | - Richard G Hunter
- Department of Psychology, University of Massachusetts, 100 Morrissey Blvd, Boston, MA, 02125, USA.
| |
Collapse
|
19
|
Svob Strac D, Vlainic J, Samardzic J, Erhardt J, Krsnik Z. Effects of acute and chronic administration of neurosteroid dehydroepiandrosterone sulfate on neuronal excitability in mice. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:1201-15. [PMID: 27051273 PMCID: PMC4807895 DOI: 10.2147/dddt.s102102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Background Neurosteroid dehydroepiandrosterone sulfate (DHEAS) has been associated with important brain functions, including neuronal survival, memory, and behavior, showing therapeutic potential in various neuropsychiatric and cognitive disorders. However, the antagonistic effects of DHEAS on γ-amino-butyric acidA receptors and its facilitatory action on glutamatergic neurotransmission might lead to enhanced brain excitability and seizures and thus limit DHEAS therapeutic applications. The aim of this study was to investigate possible age and sex differences in the neuronal excitability of the mice following acute and chronic DHEAS administration. Methods DHEAS was administered intraperitoneally in male and female adult and old mice either acutely or repeatedly once daily for 4 weeks in a 10 mg/kg dose. To investigate the potential proconvulsant properties of DHEAS, we studied the effects of acute and chronic DHEAS treatment on picrotoxin-, pentylentetrazole-, and N-methyl-D-aspartate-induced seizures in mice. The effects of acute and chronic DHEAS administration on the locomotor activity, motor coordination, and body weight of the mice were also studied. We also investigated the effects of DHEAS treatment on [3H]flunitrazepam binding to the mouse brain membranes. Results DHEAS did not modify the locomotor activity, motor coordination, body weight, and brain [3H]flunitrazepam binding of male and female mice. The results failed to demonstrate significant effects of single- and long-term DHEAS treatment on the convulsive susceptibility in both adult and aged mice of both sexes. However, small but significant changes regarding sex differences in the susceptibility to seizures were observed following DHEAS administration to mice. Conclusion Although our findings suggest that DHEAS treatment might be safe for various potential therapeutic applications in adult as well as in old age, they also support subtle interaction of DHEAS with male and female hormonal status, which may underline observed sex differences in the relationship between DHEAS and various health outcomes.
Collapse
Affiliation(s)
- Dubravka Svob Strac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Josipa Vlainic
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Janko Samardzic
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Julija Erhardt
- Department of Animal Physiology, Faculty of Science, University of Zagreb
| | - Zeljka Krsnik
- Croatian Institute for Brain Research, Department of Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
20
|
Araki R, Nishida S, Hiraki Y, Matsumoto K, Yabe T. DNA methylation of the GC box in the promoter region mediates isolation rearing-induced suppression of srd5a1 transcription in the prefrontal cortex. Neurosci Lett 2015; 606:135-9. [DOI: 10.1016/j.neulet.2015.08.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 07/30/2015] [Accepted: 08/19/2015] [Indexed: 01/01/2023]
|
21
|
Erbay LG, Kartalci S. Neurosteroid Levels in Patients with Obsessive-Compulsive Disorder. Psychiatry Investig 2015; 12:538-44. [PMID: 26508966 PMCID: PMC4620312 DOI: 10.4306/pi.2015.12.4.538] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 01/26/2015] [Accepted: 02/24/2015] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVE Changes in serum neurosteroid levels have been reported in stress-related disorders such as anxiety and depression, but not in patients with obsessive-compulsive disorder (OCD). We thus investigated such changes in patients with OCD. METHODS We compared the serum levels of progesterone, pregnanolone, dehydroepiandrosterone (DHEA), dehydroepiandrosterone sulphate (DHEA-S), cortisol and testosterone in 30 patients with OCD and 30 healthy controls. RESULTS When male and female patients were evaluated together, DHEA and cortisol levels were significantly higher in patients with OCD than the control group. When the genders were evaluated separately, DHEA and cortisol levels were higher in female patients than the female controls. The increase in DHEA levels in female patients is likely an effect of the hypothalamic-pituitary-adrenal (HPA) axis. In contrast, cortisol levels in male patients were higher than the control group, while testosterone levels were lower. The increased cortisol and decreased testosterone levels in male patients likely involves the hypothalamic-pituitary-gonadal (HPG) axis. CONCLUSION These findings suggest that neurosteroid levels in patients with OCD should be investigated together with the HPA and HPG axes in future studies.
Collapse
Affiliation(s)
| | - Sukru Kartalci
- Department of Psychiatry, Faculty of Medicine, Inonu University, Malatya, Turkey
| |
Collapse
|
22
|
Masdrakis VG, Markianos M, Oulis P. Lack of specific association between panicogenic properties of caffeine and HPA-axis activation. A placebo-controlled study of caffeine challenge in patients with panic disorder. Psychiatry Res 2015; 229:75-81. [PMID: 26243374 DOI: 10.1016/j.psychres.2015.07.069] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 06/20/2015] [Accepted: 07/26/2015] [Indexed: 12/30/2022]
Abstract
A subgroup of patients with Panic Disorder (PD) exhibits increased sensitivity to caffeine administration. However, the association between caffeine-induced panic attacks and post-caffeine hypothalamic-pituitary-adrenal (HPA)-axis activation in PD patients remains unclear. In a randomized, double-blind, cross-over experiment, 19 PD patients underwent a 400-mg caffeine-challenge and a placebo-challenge, both administered in the form of instant coffee. Plasma levels of adrenocorticotropic hormone (ACTH), cortisol and dehydroepiandrosterone sulfate (DHEAS) were assessed at both baseline and post-challenge. No patient panicked after placebo-challenge, while nine patients (47.3%) panicked after caffeine-challenge. Placebo administration did not result in any significant change in hormones' plasma levels. Overall, sample's patients demonstrated significant increases in ACTH, cortisol, and DHEAS plasma levels after caffeine administration. However, post-caffeine panickers and non-panickers did not differ with respect to the magnitude of the increases. Our results indicate that in PD patients, caffeine-induced panic attacks are not specifically associated with HPA-axis activation, as this is reflected in post-caffeine increases in ACTH, cortisol and DHEAS plasma levels, suggesting that caffeine-induced panic attacks in PD patients are not specifically mediated by the biological processes underlying fear or stress. More generally, our results add to the evidence that HPA-axis activation is not a specific characteristic of panic.
Collapse
Affiliation(s)
- Vasilios G Masdrakis
- 1st Department of Psychiatry, Athens University Medical School, Eginition Hospital, 74 Vas. Sofias Avenue, Athens 11528, Greece.
| | - Manolis Markianos
- 1st Department of Psychiatry, Athens University Medical School, Eginition Hospital, 74 Vas. Sofias Avenue, Athens 11528, Greece
| | - Panagiotis Oulis
- 1st Department of Psychiatry, Athens University Medical School, Eginition Hospital, 74 Vas. Sofias Avenue, Athens 11528, Greece
| |
Collapse
|
23
|
MacKenzie G, Maguire J. Neurosteroids and GABAergic signaling in health and disease. Biomol Concepts 2015; 4:29-42. [PMID: 25436563 DOI: 10.1515/bmc-2012-0033] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 10/12/2012] [Indexed: 11/15/2022] Open
Abstract
Endogenous neurosteroids such as allopregnanolone, allotetrahydrodeoxycorticosterone, and androstanediol are synthesized either de novo in the brain from cholesterol or are generated from the local metabolism of peripherally derived progesterone or corticosterone. Fluctuations in neurosteroid concentrations are important in the regulation of a number of physiological responses including anxiety and stress, reproductive, and sexual behaviors. These effects are mediated in part by the direct binding of neurosteroids to γ-aminobutyric acid type-A receptors (GABAARs), resulting in the potentiation of GABAAR-mediated currents. Extrasynaptic GABAARs containing the δ subunit, which contribute to the tonic conductance, are particularly sensitive to low nanomolar concentrations of neurosteroids and are likely their preferential target. Considering the large charge transfer generated by these persistently open channels, even subtle changes in neurosteroid concentrations can have a major impact on neuronal excitability. Consequently, aberrant levels of neurosteroids have been implicated in numerous disorders, including, but not limited to, anxiety, neurodegenerative diseases, alcohol abuse, epilepsy, and depression. Here we review the modulation of GABAAR by neurosteroids and the consequences for health and disease.
Collapse
|
24
|
Abstract
Neurosteroids, like allopregnanolone and pregnanolone, are endogenous regulators of neuronal excitability. Inside the brain, they are highly selective and potent modulators of GABAA receptor activity. Their anticonvulsant, anesthetics and anxiolytic properties are useful for the treatments of several neurological and psychiatric disorders via reducing the risks of side effects obtained with the commercial drugs. The principal disadvantages of endogenous neurosteroids administration are their rapid metabolism and their low oral bioavailability. Synthetic steroids analogues with major stability or endogenous neurosteroids stimulation synthesis might constitute promising novel strategies for the treatment of several disorders. Numerous studies indicate that the 3α-hydroxyl configuration is the key for binding and activity, but modifications in the steroid nucleus may emphasize different pharmacophores. So far, several synthetic steroids have been developed with successful neurosteroid-like effects. In this work, we summarize the properties of various synthetic steroids probed in trials throughout the analysis of several neurosteroids-like actions.
Collapse
Affiliation(s)
- Mariana Rey
- Laboratorio de Neurobiología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, (C1428ADN) Ciudad Autónoma de Buenos Aires, Argentina
| | - Héctor Coirini
- Laboratorio de Neurobiología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, (C1428ADN) Ciudad Autónoma de Buenos Aires, Argentina ; Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
25
|
do Rego JL, Vaudry D, Vaudry H. The non-benzodiazepine anxiolytic drug etifoxine causes a rapid, receptor-independent stimulation of neurosteroid biosynthesis. PLoS One 2015; 10:e0120473. [PMID: 25785994 PMCID: PMC4364751 DOI: 10.1371/journal.pone.0120473] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/23/2015] [Indexed: 11/19/2022] Open
Abstract
Neurosteroids can modulate the activity of the GABAA receptors, and thus affect anxiety-like behaviors. The non-benzodiazepine anxiolytic compound etifoxine has been shown to increase neurosteroid concentrations in brain tissue but the mode of action of etifoxine on neurosteroid formation has not yet been elucidated. In the present study, we have thus investigated the effect and the mechanism of action of etifoxine on neurosteroid biosynthesis using the frog hypothalamus as an experimental model. Exposure of frog hypothalamic explants to graded concentrations of etifoxine produced a dose-dependent increase in the biosynthesis of 17-hydroxypregnenolone, dehydroepiandrosterone, progesterone and tetrahydroprogesterone, associated with a decrease in the production of dihydroprogesterone. Time-course experiments revealed that a 15-min incubation of hypothalamic explants with etifoxine was sufficient to induce a robust increase in neurosteroid synthesis, suggesting that etifoxine activates steroidogenic enzymes at a post-translational level. Etifoxine-evoked neurosteroid biosynthesis was not affected by the central-type benzodiazepine (CBR) receptor antagonist flumazenil, the translocator protein (TSPO) antagonist PK11195 or the GABAA receptor antagonist bicuculline. In addition, the stimulatory effects of etifoxine and the triakontatetraneuropeptide TTN, a TSPO agonist, were additive, indicating that these two compounds act through distinct mechanisms. Etifoxine also induced a rapid stimulation of neurosteroid biosynthesis from frog hypothalamus homogenates, a preparation in which membrane receptor signalling is disrupted. In conclusion, the present study demonstrates that etifoxine stimulates neurosteroid production through a membrane receptor-independent mechanism.
Collapse
Affiliation(s)
- Jean Luc do Rego
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, Mont-Saint-Aignan, France
- Regional Platform for Cell Imaging (PRIMACEN), International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France
| | - David Vaudry
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, Mont-Saint-Aignan, France
- Regional Platform for Cell Imaging (PRIMACEN), International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France
- Neurotrophic Factors and Neuronal Differentiation team, Inserm U982, University of Rouen, Mont-Saint-Aignan, France
| | - Hubert Vaudry
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, Mont-Saint-Aignan, France
- Regional Platform for Cell Imaging (PRIMACEN), International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France
- Neurotrophic Factors and Neuronal Differentiation team, Inserm U982, University of Rouen, Mont-Saint-Aignan, France
| |
Collapse
|
26
|
Streck EL, Gonçalves CL, Furlanetto CB, Scaini G, Dal-Pizzol F, Quevedo J. Mitochondria and the central nervous system: searching for a pathophysiological basis of psychiatric disorders. REVISTA BRASILEIRA DE PSIQUIATRIA 2014; 36:156-67. [DOI: 10.1590/1516-4446-2013-1224] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 10/03/2013] [Indexed: 12/20/2022]
Affiliation(s)
- Emilio L. Streck
- Universidade do Extremo Sul Catarinense (UNESC), Brazil; National Science and Technology Institute for Translational Medicine (INCT-TM), Brazil; Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Brazil
| | - Cinara L. Gonçalves
- Universidade do Extremo Sul Catarinense (UNESC), Brazil; National Science and Technology Institute for Translational Medicine (INCT-TM), Brazil; Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Brazil
| | - Camila B. Furlanetto
- Universidade do Extremo Sul Catarinense (UNESC), Brazil; National Science and Technology Institute for Translational Medicine (INCT-TM), Brazil; Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Brazil
| | - Giselli Scaini
- Universidade do Extremo Sul Catarinense (UNESC), Brazil; National Science and Technology Institute for Translational Medicine (INCT-TM), Brazil; Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Brazil
| | - Felipe Dal-Pizzol
- Universidade do Extremo Sul Catarinense (UNESC), Brazil; National Science and Technology Institute for Translational Medicine (INCT-TM), Brazil; Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Brazil
| | - João Quevedo
- National Science and Technology Institute for Translational Medicine (INCT-TM), Brazil; Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Brazil; UNESC, Brazil
| |
Collapse
|
27
|
Bristot G, Ascoli B, Gubert C, Panizzutti B, Kapczinski F, Rosa AR. Progesterone and its metabolites as therapeutic targets in psychiatric disorders. Expert Opin Ther Targets 2014; 18:679-90. [PMID: 24654651 DOI: 10.1517/14728222.2014.897329] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Neurosteroids are molecules that regulate physiological functions of the CNS. There is increasing evidence suggesting that impaired neurosteroid biosynthesis has been associated with distinct psychiatric disorders. This review summarizes data from studies that have investigated the relationship between progesterone (PROG) and psychiatric disorders as well as the mechanisms potentially involved in PROG-induced neuroprotection. AREAS COVERED The review covers the role of PROG and its metabolites in psychiatric disorders, focusing on results from preclinical and some clinical studies that support the relationship between alterations on PROG levels and pathophysiology of psychiatric illness. We also discussed the main mechanisms underlying the neuroprotective effects of PROG metabolites. EXPERT OPINION Our review points out the possible relationship between PROG and its metabolites and the pathophysiology of psychiatric disorders. Furthermore, both preclinical and clinical studies show that certain treatments (antidepressants or antipsychotics) may normalize the levels of PROG, suggesting that the amelioration of psychiatric symptoms may occur due to upregulation of PROG metabolites. Therefore, these results give support to new possibilities of treatment for patients with psychiatric symptoms from anxiety- and depressive-like behaviors to aggressive behaviors.
Collapse
Affiliation(s)
- Giovana Bristot
- Universidade Federal do Rio Grande do Sul, National Science and Technology Institute for Translational Medicine (INCT-TM), Hospital de Clínicas de Porto Alegre, Laboratory of Molecular Psychiatry, Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) , Porto Alegre , Brazil +55 51 33598845 ; +55 51 33598846 ;
| | | | | | | | | | | |
Collapse
|
28
|
Wali B, Ishrat T, Won S, Stein DG, Sayeed I. Progesterone in experimental permanent stroke: a dose-response and therapeutic time-window study. Brain 2014; 137:486-502. [PMID: 24374329 PMCID: PMC3914469 DOI: 10.1093/brain/awt319] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/10/2013] [Accepted: 09/23/2013] [Indexed: 11/14/2022] Open
Abstract
Currently, the only approved treatment for ischaemic stroke is tissue plasminogen activator, a clot-buster. This treatment can have dangerous consequences if not given within the first 4 h after stroke. Our group and others have shown progesterone to be beneficial in preclinical studies of stroke, but a progesterone dose-response and time-window study is lacking. We tested male Sprague-Dawley rats (12 months old) with permanent middle cerebral artery occlusion or sham operations on multiple measures of sensory, motor and cognitive performance. For the dose-response study, animals received intraperitoneal injections of progesterone (8, 16 or 32 mg/kg) at 1 h post-occlusion, and subcutaneous injections at 6 h and then once every 24 h for 7 days. For the time-window study, the optimal dose of progesterone was given starting at 3, 6 or 24 h post-stroke. Behavioural recovery was evaluated at repeated intervals. Rats were killed at 22 days post-stroke and brains extracted for evaluation of infarct volume. Both 8 and 16 mg/kg doses of progesterone produced attenuation of infarct volume compared with the placebo, and improved functional outcomes up to 3 weeks after stroke on locomotor activity, grip strength, sensory neglect, gait impairment, motor coordination and spatial navigation tests. In the time-window study, the progesterone group exhibited substantial neuroprotection as late as 6 h after stroke onset. Compared with placebo, progesterone showed a significant reduction in infarct size with 3- and 6-h delays. Moderate doses (8 and 16 mg/kg) of progesterone reduced infarct size and improved functional deficits in our clinically relevant model of stroke. The 8 mg/kg dose was optimal in improving motor, sensory and memory function, and this effect was observed over a large therapeutic time window. Progesterone shows promise as a potential therapeutic agent and should be examined for safety and efficacy in a clinical trial for ischaemic stroke.
Collapse
Affiliation(s)
- Bushra Wali
- Department of Emergency Medicine, Brain Research Laboratory, Emory University, Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
29
|
Berretti R, Santoru F, Locci A, Sogliano C, Calza A, Choleris E, Porcu P, Concas A. Neonatal exposure to estradiol decreases hypothalamic allopregnanolone concentrations and alters agonistic and sexual but not affective behavior in adult female rats. Horm Behav 2014; 65:142-53. [PMID: 24368289 DOI: 10.1016/j.yhbeh.2013.12.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 12/03/2013] [Accepted: 12/15/2013] [Indexed: 01/22/2023]
Abstract
Exposure of developing female rats to estradiol during the perinatal period induced long-lasting dysregulation of gonadal axis and decreased cerebrocortical and plasma concentrations of allopregnanolone. We have now examined the effects of neonatal estradiol administration in female rats on hypothalamic allopregnanolone concentrations and on exploratory, affective, agonistic and sexual behaviors as well as social learning. A single administration of β-estradiol 3-benzoate (EB, 10μg) on the day of birth resulted in a delay of vaginal opening, acyclicity and ovarian failure. These alterations were associated with a significant decrease in the concentrations of allopregnanolone in the hypothalamus at 21 and 60days, but not at 7days, after birth. Neonatal administration of EB also increased agonistic behaviors in adult rats, such as dominant behaviors and following of an ovariectomized intruder, while living attacks unaffected. EB-treated rats showed also an increase in anogenital investigation, associated with a drastic reduction in spontaneous and induced female sexual behaviors (receptivity and proceptivity). In contrast, neonatal administration of EB did not affect locomotor activity, anxiety- and mood-related behaviors, the social transmission of flavor preferences, and seizures sensitivity. These effects of estradiol suggest that it plays a major role in regulation of both the abundance of allopregnanolone and the expression of agonistic and sexual behaviors, while failing to influence affective behaviors and social learning. Thus, the pronounced and persistent decrease in hypothalamic allopregnanolone concentration may be related to the manifestation of agonistic and sexual behaviors.
Collapse
Affiliation(s)
- R Berretti
- Department of Life and Environment Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - F Santoru
- Department of Life and Environment Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - A Locci
- Department of Life and Environment Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - C Sogliano
- Department of Life and Environment Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - A Calza
- Department of Life and Environment Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - E Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - P Porcu
- Institute of Neuroscience, National Research Council of Italy (CNR), Cagliari, Italy
| | - A Concas
- Department of Life and Environment Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy; Institute of Neuroscience, National Research Council of Italy (CNR), Cagliari, Italy.
| |
Collapse
|
30
|
Le Mellédo JM, Baker G. Role of progesterone and other neuroactive steroids in anxiety disorders. Expert Rev Neurother 2014; 4:851-60. [PMID: 15853511 DOI: 10.1586/14737175.4.5.851] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
It remains unexplained why a greater prevalence of anxiety disorders exists in women than in men, and how female hormone-related events (i.e., menstrual cycle and postpartum) can influence the course of anxiety disorders. It would appear logical that female hormones and their derivatives play a major role in these observations. The abundance of preclinical data demonstrating a role for sex hormones and their derivatives in anxiety-like behavior is in contrast to the relative paucity of experimental clinical data on the role of female hormones and neuroactive steroids in anxiety disorders. There is a dramatic potential for therapeutic anxiolytic activity of pharmacological compounds derived from powerful anxiolytic agents, such as the progesterone derivative, allopregnanolone. As a result, there is currently tremendous interest from the pharmaceutical industry in developing and testing such agents in anxiety disorders.
Collapse
|
31
|
The role of allopregnanolone in depression and anxiety. Prog Neurobiol 2013; 113:79-87. [PMID: 24215796 DOI: 10.1016/j.pneurobio.2013.09.003] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 09/21/2013] [Accepted: 09/21/2013] [Indexed: 12/22/2022]
Abstract
Neuroactive steroids such as allopregnanolone do not only act as transcriptional factors in the regulation of gene expression after intracellular back-oxidation into the 5-α pregnane steroids but may also alter neuronal excitability through interactions with specific neurotransmitter receptors. In particular, certain 3α-reduced metabolites of progesterone such as 3α,5α-tetrahydroprogesterone (allopregnanolone) and 3α,5β-tetrahydroprogesterone (pregnanolone) are potent positive allosteric modulators of the GABA(A) receptor complex. During the last years, the downregulation of neurosteroid biosynthesis has been intensively discussed to be a possible contributor to the development of anxiety and depressive disorder. Reduced levels of allopregnanolone in the peripheral blood or cerebrospinal fluid were found to be associated with major depression, anxiety disorders, premenstrual dysphoric disorder, negative symptoms in schizophrenia, or impulsive aggression. The importance of allopregnanolone for the regulation of emotion and its therapeutical use in depression and anxiety may not only involve GABAergic mechanisms, but probably also includes enhancement of neurogenesis, myelination, neuroprotection, and regulatory effects on HPA axis function. Certain pharmacokinetic obstacles limit the therapeutic use of natural neurosteroids (low bioavailability, oxidation to the ketone). Until now synthetic neuroactive steroids could not be established in the treatment of anxiety disorders or depression. However, the translocator protein (18 kDa) (TSPO) which is important for neurosteroidogenesis has been identified as a potential novel target. TSPO ligands such as XBD 173 increase neurosteroidogenesis and have anxiolytic effects with a favorable side effect profile.
Collapse
|
32
|
Carver CM, Reddy DS. Neurosteroid interactions with synaptic and extrasynaptic GABA(A) receptors: regulation of subunit plasticity, phasic and tonic inhibition, and neuronal network excitability. Psychopharmacology (Berl) 2013; 230:151-88. [PMID: 24071826 PMCID: PMC3832254 DOI: 10.1007/s00213-013-3276-5] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 08/29/2013] [Indexed: 12/25/2022]
Abstract
RATIONALE Neurosteroids are steroids synthesized within the brain with rapid effects on neuronal excitability. Allopregnanolone, allotetrahydrodeoxycorticosterone, and androstanediol are three widely explored prototype endogenous neurosteroids. They have very different targets and functions compared to conventional steroid hormones. Neuronal γ-aminobutyric acid (GABA) type A (GABA(A)) receptors are one of the prime molecular targets of neurosteroids. OBJECTIVE This review provides a critical appraisal of recent advances in the pharmacology of endogenous neurosteroids that interact with GABA(A) receptors in the brain. Neurosteroids possess distinct, characteristic effects on the membrane potential and current conductance of the neuron, mainly via potentiation of GABA(A) receptors at low concentrations and direct activation of receptor chloride channel at higher concentrations. The GABA(A) receptor mediates two types of inhibition, now characterized as synaptic (phasic) and extrasynaptic (tonic) inhibition. Synaptic release of GABA results in the activation of low-affinity γ2-containing synaptic receptors, while high-affinity δ-containing extrasynaptic receptors are persistently activated by the ambient GABA present in the extracellular fluid. Neurosteroids are potent positive allosteric modulators of synaptic and extrasynaptic GABA(A) receptors and therefore enhance both phasic and tonic inhibition. Tonic inhibition is specifically more sensitive to neurosteroids. The resulting tonic conductance generates a form of shunting inhibition that controls neuronal network excitability, seizure susceptibility, and behavior. CONCLUSION The growing understanding of the mechanisms of neurosteroid regulation of the structure and function of the synaptic and extrasynaptic GABA(A) receptors provides many opportunities to create improved therapies for sleep, anxiety, stress, epilepsy, and other neuropsychiatric conditions.
Collapse
Affiliation(s)
- Chase Matthew Carver
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, 2008 Medical Research and Education Building, 8447 State Highway 47, Bryan, TX, 77807-3260, USA
| | | |
Collapse
|
33
|
Plescia F, Sardo P, Rizzo V, Cacace S, Marino RAM, Brancato A, Ferraro G, Carletti F, Cannizzaro C. Pregnenolone sulphate enhances spatial orientation and object discrimination in adult male rats: evidence from a behavioural and electrophysiological study. Behav Brain Res 2013; 258:193-201. [PMID: 24149069 DOI: 10.1016/j.bbr.2013.10.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 10/08/2013] [Accepted: 10/14/2013] [Indexed: 11/16/2022]
Abstract
Neurosteroids can alter neuronal excitability interacting with specific neurotransmitter receptors, thus affecting several functions such as cognition and emotionality. In this study we investigated, in adult male rats, the effects of the acute administration of pregnenolone-sulfate (PREGS) (10mg/kg, s.c.) on cognitive processes using the Can test, a non aversive spatial/visual task which allows the assessment of both spatial orientation-acquisition and object discrimination in a simple and in a complex version of the visual task. Electrophysiological recordings were also performed in vivo, after acute PREGS systemic administration in order to investigate on the neuronal activation in the hippocampus and the perirhinal cortex. Our results indicate that, PREGS induces an improvement in spatial orientation-acquisition and in object discrimination in the simple and in the complex visual task; the behavioural responses were also confirmed by electrophysiological recordings showing a potentiation in the neuronal activity of the hippocampus and the perirhinal cortex. In conclusion, this study demonstrates that PREGS systemic administration in rats exerts cognitive enhancing properties which involve both the acquisition and utilization of spatial information, and object discrimination memory, and also correlates the behavioural potentiation observed to an increase in the neuronal firing of discrete cerebral areas critical for spatial learning and object recognition. This provides further evidence in support of the role of PREGS in exerting a protective and enhancing role on human memory.
Collapse
Affiliation(s)
- Fulvio Plescia
- Department of Sciences for Health Promotion and Mother and Child Care "Giuseppe D'Alessandro", University of Palermo, V. Vespro 129, 90127 Palermo, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Olayiwola G, Ukponmwan O, Olawode D. Sedative and anxiolytic effects of the extracts of the leaves of Stachytarpheta cayennensis in mice. AFRICAN JOURNAL OF TRADITIONAL, COMPLEMENTARY, AND ALTERNATIVE MEDICINES : AJTCAM 2013; 10:568-79. [PMID: 24311890 PMCID: PMC3847405 DOI: 10.4314/ajtcam.v10i6.32] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The leaves are used ethnomedicinally in Nigeria and other parts of the world for insomnia and anxiety among other uses. The investigations sought scientific evidence for the ethnomedicinal use of the leaves for the management of insomnia and anxiety as well as the neural mechanisms for the activities. The sedative and anxiolytic effects of the extracts of the leaves of Stachytarpheta cayennensis were examined in this study. The methanolic extract (5-50 mg/kg, i.p.) as well as the ethylacetate (10-50 mg/kg, i.p.), butanol and aqueous fractions (5-50 mg/kg, i.p.) of the extract were examined. Sedation was assessed as reduced novelty-induced rearing (NIR), reduced spontaneous locomotor activity (SLA) and increased pentobarbitone-induced sleeping time (PIST) in mice. The anti-anxiety effect (methanol 2.5-5.0; butanol 5.0; aqueous 20.0; ethylacetate 25.0 mg/kg, i.p.) was assessed using an elevated plus maze. LD50 was calculated for the extract and the fractions after the intraperitoneal route of administration using the Locke method. The methanolic extract, the butanol and the aqueous fractions inhibited rearing and spontaneous locomotion but prolonged pentobarbitone induced sleep. The ethylacetate fraction however increased both rearing and locomotion and decreased pentobarbitone sleeping time. The butanol and aqueous fractions, but not the methanol extract showed indices of open arm avoidance consistent with anti-anxiety effect. Naltrexone (2.5 mg/kg, i.p.) reversed the inhibition of rearing, locomotion and prolongation of pentobarbitone sleep due to the aqueous fraction of the extract. Flumazenil (2mg/kg, i.p.) abolished the effects of both methanolic extract and the butanol fraction on rearing, locomotion, pentobarbitone sleep and anxiety model. The methanolic extract, the butanol and aqueous fractions possess sedative activity while the ethylacetate fraction possesses stimulant property. The anxiolytic effect was found in both the aqueous fraction and the butanol fraction but not in the main methanol extract and also not in the ethylacetate fraction. Flumazenil, blocked the effect of the leaves of Stachytarpheta cayennensis on rearing, locomotion and elevated plus maze suggesting that GABA receptors are involved in the observed sedative and anxiolytic activities. This study also found opioid receptors involved in the sedative activity of the leaves of Stachytarpheta cayennensis. The rationale for the ethnomedicinal use of the leaves for the management of insomnia and anxiety were confirmed scientifically in this study.
Collapse
Affiliation(s)
- Gbola Olayiwola
- Department of Clinical Pharmacy and Pharmacy Administration, Obafemi Awolowo University, Ile-Ife, Nigeria
| | | | | |
Collapse
|
35
|
Koonce CJ, Frye CA. Progesterone facilitates exploration, affective and social behaviors among wildtype, but not 5α-reductase Type 1 mutant, mice. Behav Brain Res 2013; 253:232-9. [PMID: 23886595 DOI: 10.1016/j.bbr.2013.07.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 07/10/2013] [Accepted: 07/15/2013] [Indexed: 12/24/2022]
Abstract
Progesterone (P4) facilitates exploration, anxiety and social behaviors in estrogen (E2)-primed mice. Some of these effects may be due to actions of its 5α-reduced metabolite, 5α-pregnan-3α-ol-20-one (3α,5α-THP). In order to address the role of P4 and its metabolite, 3α,5α-THP, a mouse model was utilized. We hypothesized that if P4's metabolism to 3α,5α-THP is essential to facilitate exploratory, anti-anxiety and social behaviors of mice, then wildtype, but not 5α-reductase knockout (5α-RKO), mice will have greater expression of these behaviors. Experiment 1: Mice were ovariectomized (ovx), E2-primed and administered P4 (0, 125, 250, or 500μg) subcutaneously and then tested 4h later in a battery of tasks: open field, elevated plus maze, and social interaction. Experiment 2: Ovx, E2-primed mice were administered P4 (4mg/kg), 3α,5α-THP (4mg/kg), medroxyprogesterone acetate (MPA, which does not convert to 3α,5α-THP; 4mg/kg), or vehicle subcutaneously and tested 4h later. There was a dose-dependent effect of P4 to wildtype, but not 5α-RKO, mice. Neither wildtype, nor 5α-RKO, mice had increased exploration, anti-anxiety or pro-social behavior with MPA administration. Progesterone only exerted effects on anti-anxiety behavior, and increased 3α,5α-THP in the prefrontal cortex and hippocampus, when administered to wildtype mice. 3α,5α-THP to both WT and 5α-RKO mice increased exploration, anti-anxiety and social interaction and 3α,5α-THP levels in the hippocampus and prefrontal cortex. Thus, metabolism of P4 by the 5α-reductase enzyme may be essential for enhancement of these behaviors.
Collapse
Affiliation(s)
- Carolyn J Koonce
- Department of Psychology, The University at Albany-SUNY, Albany, NY 12222, USA
| | | |
Collapse
|
36
|
Plescia F, Marino RAM, Cannizzaro E, Brancato A, Cannizzaro C. The role of pregnenolone sulphate in spatial orientation-acquisition and retention: an interplay between cognitive potentiation and mood regulation. Behav Processes 2013; 99:130-7. [PMID: 23860279 DOI: 10.1016/j.beproc.2013.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 07/04/2013] [Accepted: 07/05/2013] [Indexed: 01/23/2023]
Abstract
Neurosteroids can alter neuronal excitability interacting with specific neurotransmitter receptors, thus affecting several functions such as cognition and emotionality. In this study, we investigated, in adult male rats, the effects of the acute administration of pregnenolone-sulfate (PREGS) (10 mg/Kg, s.c.) on cognitive processes using the Can test, a non aversive spatial/visual task which allows the assessment of spatial information-acquisition during the baseline training, and of memory retention in the longitudinal study. Furthermore, on the basis of PREGS pharmacological profile, the modulation of depressive-like behaviour was also evaluated in the forced swim test (FST). Our results indicate that acute PREGS induces: an improvement in spatial orientation-acquisition and in reference memory, during the baseline training; a strengthening effect on reference and working memory during the longitudinal study. A decrease in immobility time in the FST has also been recorded. In conclusion, PREGS exerts enhancing properties on acquisition, consolidation and retrieval of spatial information, probably due of improved hippocampal-dependent memory processes. The additional antidepressant effect observed in the FST can provide further evidence in support of the potential of PREGS as a therapeutic tool for the treatment of cognitive deficits associated with mood disorders. This article is part of a Special Issue entitled: insert SI title.
Collapse
Affiliation(s)
- Fulvio Plescia
- Department of Sciences for Health Promotion and Mother and Child Care "Giuseppe D'Alessandro", University of Palermo, V. Vespro 129, 90127 Palermo, Italy
| | | | | | | | | |
Collapse
|
37
|
|
38
|
Alteration of neonatal Allopregnanolone levels affects exploration, anxiety, aversive learning and adult behavioural response to intrahippocampal neurosteroids. Behav Brain Res 2012; 241:96-104. [PMID: 23228522 DOI: 10.1016/j.bbr.2012.11.043] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 11/28/2012] [Accepted: 11/29/2012] [Indexed: 11/20/2022]
Abstract
Neurosteroids (NS) are well known to exert modulatory effects on ionotropic receptors. Recent findings indicate that NS could also act as important factors during development. In this sense, neonatal modifications of Allopregnanolone (Allop) levels during critical periods have been demonstrate to alter the morphology of the hippocampus but also other brain structures. The aim of the present work is to screen whether the alterations of Allop levels modify adult CA1 hippocampal response to NS administration. For this purpose, pups were injected with Allop (20 mg/kg s.c.), Finasteride (5α-reductase inhibitor that impedes Allop synthesis) (50 mg/kg s.c.) or Vehicle from postnatal day 5 (P5) to postnatal day 9 (P9). NS levels were tested at P5. To test the behavioural hippocampal response to NS in adulthood, animals were implanted with a bilateral cannula into the CA1 hippocampus at 80 days old and injected with Allop (0.2 μg/0.5 μl), Pregnenolone sulphate (5 ng/0.5 μl) or Vehicle in each hippocampus. After injections animals were tested in the Boisser test to assess exploratory behaviour, the elevated plus maze to assess anxiety and the passive avoidance to test aversive learning. Results indicate that alteration of neonatal Allop or pregnenolone levels (by Allop and Finasteride administration, respectively) suppressed intrahippocampal Allop anxiolytic effect in the EPM. Moreover our results also indicate that manipulation of neonatal Allop levels (Allop and Finast administration) alters exploratory and anxiety-like behaviour and impairs aversive learning in the adulthood. These data point out the role of Allop in the maturation of hippocampal function and behaviour.
Collapse
|
39
|
Circadian rhythms in obsessive–compulsive disorder. J Neural Transm (Vienna) 2012; 119:1077-83. [DOI: 10.1007/s00702-012-0805-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 04/12/2012] [Indexed: 11/25/2022]
|
40
|
Neurosteroidogenesis is required for the physiological response to stress: role of neurosteroid-sensitive GABAA receptors. J Neurosci 2012; 31:18198-210. [PMID: 22171026 DOI: 10.1523/jneurosci.2560-11.2011] [Citation(s) in RCA: 197] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis, which mediates the body's response to stress, is largely under GABAergic control. Here we demonstrate that corticotropin-releasing hormone (CRH) neurons are modulated by the stress-derived neurosteroid, tetrahydrodeoxycorticosterone (THDOC), acting on δ subunit-containing GABA(A) receptors (GABA(A)Rs). Under normal conditions, THDOC potentiates the inhibitory effects of GABA on CRH neurons, decreasing the activity of the HPA axis. Counterintuitively, following stress, THDOC activates the HPA axis due to dephosphorylation of KCC2 residue Ser940, resulting in a collapse of the chloride gradient and excitatory GABAergic transmission. The effects of THDOC on CRH neurons are mediated by actions on GABA(A)R δ subunit-containing receptors since these effects are abolished in Gabrd(-/-) mice under both control and stress conditions. Interestingly, blocking neurosteroidogenesis with finasteride is sufficient to block the stress-induced elevations in corticosterone and prevent stress-induced anxiety-like behaviors in mice. These data demonstrate that positive feedback of neurosteroids onto CRH neurons is required to mount the physiological response to stress. Further, GABA(A)R δ subunit-containing receptors and phosphorylation of KCC2 residue Ser940 may be novel targets for control of the stress response, which has therapeutic potential for numerous disorders associated with hyperexcitability of the HPA axis, including Cushing's syndrome, epilepsy, and major depression.
Collapse
|
41
|
Korinek M, Kapras V, Vyklicky V, Adamusova E, Borovska J, Vales K, Stuchlik A, Horak M, Chodounska H, Vyklicky L. Neurosteroid modulation of N-methyl-D-aspartate receptors: molecular mechanism and behavioral effects. Steroids 2011; 76:1409-18. [PMID: 21925193 DOI: 10.1016/j.steroids.2011.09.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 08/31/2011] [Accepted: 09/01/2011] [Indexed: 12/12/2022]
Abstract
Glutamate is the main neurotransmitter released at synapses in the central nervous system of vertebrates. Its excitatory role is mediated through activation of specific glutamatergic ionotropic receptors, among which the N-methyl-D-aspartate (NMDA) receptor subtype has attracted considerable attention in recent years. Substantial progress has been made in elucidating the roles these receptors play under physiological and pathological conditions and in our understanding of the functional, structural, and pharmacological properties of NMDA receptors. Many pharmacological compounds have been identified that affect the activity of NMDA receptors, including neurosteroids. This review summarizes our knowledge about molecular mechanisms underlying the neurosteroid action at NMDA receptors as well as about the action of neurosteroids in animal models of human diseases.
Collapse
Affiliation(s)
- Miloslav Korinek
- Institute of Physiology, Academy of Sciences of the Czech Republic, vvi Videnska 1083, 142 20 Prague 4, Czech Republic
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Childs E, O'Connor S, de Wit H. Bidirectional interactions between acute psychosocial stress and acute intravenous alcohol in healthy men. Alcohol Clin Exp Res 2011; 35:1794-803. [PMID: 21762177 DOI: 10.1111/j.1530-0277.2011.01522.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND The biological mechanisms by which acute stress increases alcohol consumption are unclear. One potential mechanism is that stress acts by altering the pharmacological and subjective effects of alcohol. Acute stress produces a cascade of physiological and psychological effects, each with a distinctive time course. In this study, we investigated whether different phases of response to an acute stress alter the subjective effects of intravenous alcohol, by administering the drug at 2 different times after the stress. METHODS Healthy men (n = 25) participated in 2 sessions: 1 with the Trier Social Stress Test and the other with a nonstressful control task, each followed by infusions of intravenous alcohol (targeting 40 mg% in 5 minutes) and placebo. One group of participants received alcohol within 1 minute of completing the tasks (Alc0, n = 11), followed by placebo 30 minutes later. In the other group (Alc30, n = 14), the order of alcohol and placebo infusions was reversed. Subjective effects (i.e., anxiety, stimulation, want more) and physiological measures (heart rate, blood pressure, salivary cortisol) were measured before and at repeated intervals after the tasks and infusions. RESULTS Stress did not change the subjective effects of alcohol in either group. However, when individual differences in alcohol responses were considered, stress differentially altered the stimulant-like and sedative effects of alcohol. Among individuals who exhibited predominantly stimulant responses to alcohol in the nonstressful condition, stress decreased the stimulant-like effects of alcohol and "wanting more." By contrast, among participants who did not report stimulation after alcohol in the control session, stress decreased the sedative effects and increased "want more." In addition, alcohol administered immediately after the Trier Social Stress Test dampened cortisol responses yet prolonged negative subjective responses to the stress. CONCLUSIONS These findings demonstrate that there are bidirectional relationships between alcohol and stress. Alcohol influences responses to stress, and stress changes reactions to alcohol, depending on an individual's pattern of response to alcohol. This study highlights the fact that stress-alcohol interactions vary among individual drinkers, suggesting that the effects of stress on motivation to drink alcohol may also differ between individuals.
Collapse
Affiliation(s)
- Emma Childs
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois 60637, USA.
| | | | | |
Collapse
|
43
|
Mòdol L, Darbra S, Pallarès M. Neurosteroids infusion into the CA1 hippocampal region on exploration, anxiety-like behaviour and aversive learning. Behav Brain Res 2011; 222:223-9. [PMID: 21463656 DOI: 10.1016/j.bbr.2011.03.058] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 03/21/2011] [Accepted: 03/27/2011] [Indexed: 12/25/2022]
Abstract
Neurosteroids (NS) are substances synthesised de novo in the brain that have rapid modulatory effects on ionotropic receptors. Specifically, NS can act as positive allosteric modulators of GABAA receptors as pregnanolone or allopregnanolone (Allop), or GABAA negative modulators and NMDA positive modulators as pregnenolone (PREG) or dehydroepiandrosterone (DHEA) and their sulphate esters (PREGS and DHEAS). Given this, their role in anxiety and emotional disturbances has been suggested. In addition, NS such as PREGS or DHEAS have demonstrated a promnesic role in several learning tests. The aim of the present work is to highlight the role that the dorsal (CA1) hippocampus plays in the behavioural profile of NS such as Allop and PREGS in tests assessing exploration, anxiety and aversive learning in rats. For this purpose, animals were administered intrahippocampally with Allop (0.2μg/0.5μl), PREGS (5ng/0.5μl) or vehicle in each hippocampus, and tested in the Boissier and elevated plus maze (EPM) tests. For learning test we have chosen the passive avoidance paradigm. Results indicate that intrahippocampal administration of Allop enhances exploration, reflected in an increase in the total and the inner number of head-dips. Allop-injected animals also showed an increase in the percentage of entries into the open arms of the EPM, suggesting an anxiolytic-like profile. In addition, post-acquisition PREGS administration enhanced passive avoidance retention, while post-acquisition Allop administration had no effects on aversive learning retention. These results point out the important role of the dorsal (CA1) hippocampus in several NS behavioural effects, such as exploration, anxiety, learning and memory.
Collapse
Affiliation(s)
- Laura Mòdol
- Departament de Psicobiologia i Metodologia en Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | | | | |
Collapse
|
44
|
Schüle C, Eser D, Baghai TC, Nothdurfter C, Kessler JS, Rupprecht R. Neuroactive steroids in affective disorders: target for novel antidepressant or anxiolytic drugs? Neuroscience 2011; 191:55-77. [PMID: 21439354 DOI: 10.1016/j.neuroscience.2011.03.025] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 03/13/2011] [Accepted: 03/14/2011] [Indexed: 11/18/2022]
Abstract
In the past decades considerable evidence has emerged that so-called neuroactive steroids do not only act as transcriptional factors in the regulation of gene expression but may also alter neuronal excitability through interactions with specific neurotransmitter receptors such as the GABA(A) receptor. In particular, 3α-reduced neuroactive steroids such as allopregnanolone or allotetrahydrodeoxycorticosterone have been shown to act as positive allosteric modulators of the GABA(A) receptor and to play an important role in the pathophysiology of depression and anxiety. During depression, the concentrations of 3α,5α-tetrahydroprogesterone and 3α,5β-tetrahydroprogesterone are decreased, while the levels of 3β,5α-tetrahydroprogesterone, a stereoisomer of 3α,5α-tetrahydroprogesterone, which may act as an antagonist for GABAergic steroids, are increased. Antidepressant drugs such as selective serotonin reuptake inhibitors (SSRIs) or mirtazapine apparently have an impact on key enzymes of neurosteroidogenesis and have been shown to normalize the disequilibrium of neuroactive steroids in depression by increasing 3α-reduced pregnane steroids and decreasing 3β,5α-tetrahydroprogesterone. Moreover, 3α-reduced neuroactive steroids have been demonstrated to possess antidepressant- and anxiolytic-like effects both in animal and human studies for themselves. In addition, the translacator protein (18 kDa) (TSPO), previously called peripheral benzodiazepine receptor, is the key element of the mitochondrial import machinery supplying the substrate cholesterol to the first steroidogenic enzyme (P450scc), which transforms cholesterol into pregnenolone, the precursor of all neurosteroids. TSPO ligands increase neurosteroidogenesis and are a target of novel anxiolytic drugs producing anxiolytic effects without causing the side effects normally associated with conventional benzodiazepines such as sedation or tolerance. This article is part of a Special Issue entitled: Neuroactive Steroids: Focus on Human Brain.
Collapse
Affiliation(s)
- C Schüle
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University, Nussbaumstrasse 7, 80336 Munich, Germany.
| | | | | | | | | | | |
Collapse
|
45
|
Longone P, di Michele F, D’Agati E, Romeo E, Pasini A, Rupprecht R. Neurosteroids as neuromodulators in the treatment of anxiety disorders. Front Endocrinol (Lausanne) 2011; 2:55. [PMID: 22654814 PMCID: PMC3356011 DOI: 10.3389/fendo.2011.00055] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 10/03/2011] [Indexed: 11/22/2022] Open
Abstract
Anxiety disorders are the most common psychiatric disorders. They are frequently treated with benzodiazepines, which are fast acting highly effective anxiolytic agents. However, their long-term use is impaired by tolerance development and abuse liability. In contrast, antidepressants such as selective serotonin reuptake inhibitors (SSRIs) are considered as first-line treatment but have a slow onset of action. Neurosteroids are powerful allosteric modulators of GABA(A) and glutamate receptors. However, they also modulate sigma receptors and they are modulated themselves by SSRIs. Both pre-clinical and clinical studies have shown that neurosteroid homeostasis is altered in depression and anxiety disorders and antidepressants may act in part through restoring neurosteroid disbalance. Moreover, novel drugs interfering with neurosteroidogenesis such as ligands of the translocator protein (18 kDa) may represent an attractive pharmacological option for novel anxiolytics which lack the unwarranted side effects of benzodiazepines. Thus, neurosteroids are important endogenous neuromodulators for the physiology and pathophysiology of anxiety and they may constitute a novel therapeutic approach in the treatment of these disorders.
Collapse
Affiliation(s)
- Patrizia Longone
- Molecular Neurobiology Unit, Experimental NeurologyFondazione Santa Lucia, Rome, Italy
- *Correspondence: Patrizia Longone, Molecular Neurobiology Unit, Room 201, Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy. e-mail: ; Rainer Rupprecht, Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053 Regensburg, Germany. e-mail:
| | | | - Elisa D’Agati
- Unit of Child Neurology and Psychiatry, Department of Neuroscience, University of Rome “Tor Vergata,”Rome, Italy
| | - Elena Romeo
- Department of Neuroscience, University of Rome “Tor Vergata,”Rome, Italy
| | - Augusto Pasini
- Unit of Child Neurology and Psychiatry, Department of Neuroscience, University of Rome “Tor Vergata,”Rome, Italy
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University RegensburgRegensburg, Germany
- *Correspondence: Patrizia Longone, Molecular Neurobiology Unit, Room 201, Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy. e-mail: ; Rainer Rupprecht, Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053 Regensburg, Germany. e-mail:
| |
Collapse
|
46
|
Nelson M, Pinna G. S-norfluoxetine microinfused into the basolateral amygdala increases allopregnanolone levels and reduces aggression in socially isolated mice. Neuropharmacology 2010; 60:1154-9. [PMID: 20971127 DOI: 10.1016/j.neuropharm.2010.10.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 10/07/2010] [Accepted: 10/14/2010] [Indexed: 01/24/2023]
Abstract
A decrease of brain allopregnanolone biosynthesis may play a role in emotion, impulsive behavior, and anxiety spectrum disorders by decreasing GABAergic neurotransmission. In male mice, four weeks of social isolation induces behavioral dysfunctions such as aggression, fear, and anxiety-like behavior associated with a decrease in allopregnanolone biosynthesis in selected corticolimbic structures comprising the basolateral amygdala (BLA), olfactory bulb, hippocampus, and medial prefrontal cortex. Importantly, no decrease in allopregnanolone biosynthesis has been found in the striatum and cerebellum. Given the importance of the amygdaloid complex in emotional behavior, we hypothesized that this brain area may play a pivotal role in decreasing social isolation-induced aggression. Thus, socially isolated mice were directly infused with S-norfluoxetine (S-NFLX) or pregnanolone (an analog of allopregnanolone) into the BLA and striatum. When S-NFLX (2.5, 3.75, and 5 nmol/0.2 μl) or pregnanolone (1.25, 2.5, 3.75, and 5.0 nmol/0.2 μl) is directly infused into the BLA, these agents dose-dependently reduced aggression (S-NFLX up to 93% and pregnanolone up to 96%) of a socially isolated mouse to a same-sex intruder. However, S-NFLX (3.75 and 5 nmol) infused directly into the striatum failed to alter aggression. Allopregnanolone content in the BLA after S-NFLX (3.75 nmol) infusion was increased by 3-fold and in the hippocampus, by 80%. Allopregnanolone levels did not change in the olfactory bulb or in the frontal cortex of the same mice. S-NFLX (3.75 nmol) infused into the striatum failed to increase the levels of allopregnanolone. These results suggest that S-NFLX, acting as a selective brain steroidogenic stimulant (SBSS), increases corticolimbic allopregnanolone levels and regulates aggression, which underscores the pivotal role of the BLA and hippocampus in the regulation of aggressiveness in socially isolated mice. This article is part of a Special Issue entitled 'Trends in neuropharmacology: in memory of Erminio Costa'.
Collapse
Affiliation(s)
- Marianela Nelson
- Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, 1601 W Taylor Street, Chicago, IL 60612, USA
| | | |
Collapse
|
47
|
Chen L, Cai W, Chen L, Zhou R, Furuya K, Sokabe M. Modulatory metaplasticity induced by pregnenolone sulfate in the rat hippocampus: a leftward shift in LTP/LTD-frequency curve. Hippocampus 2010; 20:499-512. [PMID: 19475651 DOI: 10.1002/hipo.20649] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
We recently have found that an acute application of the neurosteroid pregnenolone sulfate (PREGS) at 50 muM to rat hippocampal slices induces a long-lasting potentiation (LLP(PREGS)) via a sustained ERK2/CREB activation at perforant-path/granule-cell synapses in the dentate gyrus. This study is a follow up to investigate whether the expression of LLP(PREGS) influences subsequent frequency-dependent synaptic plasticity. Conditioning electric stimuli (CS) at 0.1-200 Hz were given to the perforant-path of rat hippocampal slices expressing LLP(PREGS) to induce long-term potentiation (LTP) and long-term depression (LTD). The largest LTP was induced at about 20 Hz-CS, which is normally a subthreshold frequency, and the largest LTD at 0.5 Hz-CS, resulting in a leftward-shift of the LTP/LTD-frequency curve. Furthermore, the level of LTP at 100 Hz-CS was significantly attenuated to give band-pass filter characteristics of LTP induction with a center frequency of about 20 Hz. The LTP induced by 20 Hz-CS (termed 20 Hz-LTP) was found to be postsynaptic origin and dependent on L-type voltage-gated calcium channel (L-VGCC) but not on N-methyl-D-aspartate receptor (NMDAr). Moreover, the induction of 20 Hz-LTP required a sustained activation of ERK2 that had been triggered by PREGS. In conclusion, the transient elevation of PREGS is suggested to induce a modulatory metaplasticity through a sustained activation of ERK2 in an L-VGCC dependent manner.
Collapse
Affiliation(s)
- Ling Chen
- Laboratory of Reproductive Medicine, Department of Physiology, Nanjing Medical University, Nanjing, China.
| | | | | | | | | | | |
Collapse
|
48
|
Kohtz AS, Paris JJ, Frye CA. Low doses of cocaine decrease, and high doses increase, anxiety-like behavior and brain progestogen levels among intact rats. Horm Behav 2010; 57:474-80. [PMID: 20171966 PMCID: PMC3608214 DOI: 10.1016/j.yhbeh.2010.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Revised: 02/03/2010] [Accepted: 02/07/2010] [Indexed: 11/20/2022]
Abstract
There are sex and hormonal differences in response to cocaine that have been demonstrated in people and animal models. Cocaine can alter secretion of progestogens, such as progesterone (P), and its neuroactive metabolite, 5alpha-pregnan-3alpha-ol-20-one (3alpha,5alpha-THP). However, little research has been done on the neuroendocrine effects in the initiation phase of cocaine use. We hypothesize that some sex/hormonal differences in initiation phase responses to cocaine may be related to formation of progestogens. To investigate the role of progestogens in sex differences in response to acute cocaine, male and female rats in the high (proestrous) or low (diestrous) progestogen phase of the estrous cycle were administered cocaine (0, 5, 10, or 20mg/kg, IP). We examined cocaine's acute neuroendocrine effects on P and 3alpha,5alpha-THP levels, as well as its effects on acute psychomotor stimulation, anxiety, and sexual behaviors. Among rats that had P and/or 3alpha,5alpha-THP levels increased in response to cocaine, enhanced acute psychomotor stimulation was observed. Results suggest that cocaine produces U-shaped curves for progestogens, and anxiety-like behaviors. Male rats were less susceptible to these effects of cocaine than were proestrous or diestrous female rats. However, cocaine's disruption of sexual behaviors was similar among males and proestrous females. These data suggest a complex interaction between hormonal milieu and the neuroendocrine and behavioral effects of cocaine.
Collapse
Affiliation(s)
- Amy S. Kohtz
- Department of Psychology, The University at Albany-SUNY, USA
| | - Jason J. Paris
- Department of Psychology, The University at Albany-SUNY, USA
| | - Cheryl A. Frye
- Department of Psychology, The University at Albany-SUNY, USA
- Department of Biological Sciences, The University at Albany-SUNY, USA
- Center for Life Sciences, The University at Albany-SUNY, USA
- Center for Neuroscience Research, The University at Albany-SUNY, USA
- Corresponding author. Department of Psychology, The University at Albany-SUNY, Life Sciences Research Building 01058, 1400 Washington Avenue, Albany, NY 12222, USA. Fax: +1 518 591 8848. (C.A. Frye)
| |
Collapse
|
49
|
Abstract
This chapter provides an overview of neurosteroids, especially their impact on the brain, sex differences and their therapeutic potentials. Neurosteroids are synthesized within the brain and rapidly modulate neuronal excitability. They are classified as pregnane neurosteroids, such as allopregnanolone and allotetrahydrodeoxycorticosterone, androstane neurosteroids, such as androstanediol and etiocholanolone, and sulfated neurosteroids such as pregnenolone sulfate. Neurosteroids such as allopregnanolone are positive allosteric modulators of GABA-A receptors with powerful anti-seizure activity in diverse animal models. Neurosteroids increase both synaptic and tonic inhibition. They are endogenous regulators of seizure susceptibility, anxiety, and stress. Sulfated neurosteroids such as pregnenolone sulfate, which are negative GABA-A receptor modulators, are memory-enhancing agents. Sex differences in susceptibility to brain disorders could be due to neurosteroids and sexual dimorphism in specific structures of the human brain. Synthetic neurosteroids that exhibit better bioavailability and efficacy and drugs that enhance neurosteroid synthesis have therapeutic potential in anxiety, epilepsy, and other brain disorders. Clinical trials with the synthetic neurosteroid analog ganaxolone in the treatment of epilepsy have been encouraging. Neurosteroidogenic agents that lack benzodiazepine-like side effects show promise in the treatment of anxiety and depression.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, College Station, TX, USA.
| |
Collapse
|
50
|
S N U, J M V, N S J, P V D. Neurosteroids modulate compulsive and persistent behavior in rodents: implications for obsessive-compulsive disorder. Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:1161-6. [PMID: 19549549 DOI: 10.1016/j.pnpbp.2009.06.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Revised: 05/28/2009] [Accepted: 06/14/2009] [Indexed: 10/20/2022]
Abstract
Neurosteroids are reported to modulate GABAergic and glutamatergic pathways that then influence serotonin and dopamine, the neurotransmitters implicated in pathophysiology of obsessive-compulsive disorder (OCD). Fluoxetine, a selective serotonin reuptake inhibitor clinically used in OCD is reported to increase the levels of neurosteroids like allopregnanolone, whereas OCD patients exhibit higher plasma levels of dehydroepiandrosterone 3-sulphate (DHEAS), a neuroactive steroid having opposite effects to that of allopregnanolone. Hence, it was contemplated that neurosteroids may influence obsessive-compulsive behavior. To test this possibility we studied the influence of various neurosteroids on two behavioral models of OCD, namely marble-burying behavior in mice and 8-OH-DPAT induced disruption of spontaneous alternation behavior (SAB) in rats. The results revealed that allopregnanolone (1 microg/mouse, i.c.v) and progesterone (20mg/kg, s.c.) reduced the marble-burying behavior in mice, whereas dehydroisoandrosterone 3-sulphate (DHAS) (5mg/kg, i.p.) exacerbated the same. The effects of allopregnanolone were comparable to that of fluoxetine (10mg/kg, i.p.). In view of the report that restraint stress increases the levels of allopregnanolone and isolation stress decreases the same, we studied the effect of these stressors on marble-burying behavior; wherein it was found to be less in restraint stress exposed mice, and higher in socially isolated mice. Restrain stress-induced attenuation of marble-burying behavior was blocked by finasteride, a neurosteroid biosynthesis blocker. In rat model of SAB disruption, acute and chronic treatment with allopregnanolone (1 microg/mouse, i.c.v.) reduced 8-OH-DPAT-induced persistent behavior, whereas treatment with DHAS (5mg/kg, i.p.) had an opposite effect. In conclusion, the studies indicate that neurosteroids can modulate obsessive-compulsive behavior in a bidirectional manner, and could serve as an effective target in the management of OCD.
Collapse
Affiliation(s)
- Umathe S N
- University Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Mahatma Jyotiba Fuley Shaikshanik Parisar, Amravati Road, Nagpur, MS 440 033, India.
| | | | | | | |
Collapse
|