1
|
Servi R, Akkoç RF, Aksu F, Servi S. Therapeutic Potential of Enzymes, Neurosteroids, and Synthetic Steroids in Neurodegenerative Disorders: A Critical Review. J Steroid Biochem Mol Biol 2025; 251:106766. [PMID: 40288591 DOI: 10.1016/j.jsbmb.2025.106766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Neurodegenerative disorders present a significant challenge to healthcare systems, mainly due to the limited availability of effective treatment options to halt or reverse disease progression. Endogenous steroids synthesized in the central nervous system, such as pregnenolone (PREG), dehydroepiandrosterone (DHEA), progesterone (PROG), and allopregnanolone (ALLO), have been identified as potential therapeutic agents for neurodegenerative diseases. Neurosteroids such as ALLO, DHEA, and PROG, as well as their synthetic analogs like Ganaxolene, Fluasterone, and Olexoxime, offer promising effects for conditions such as Alzheimer's disease (AD) and depression. Moreover, Brexanolone and Ganaxolone are synthetic steroids approved for the treatment of postpartum depression and epilepsy, respectively. Neurosteroids such as ALLO are crucial in modulating GABAergic neurotransmission and reducing neuroinflammation. These compounds enhance the activity of GABA-A receptors, leading to increased inhibitory signaling in the brain, which can help regulate mood, cognition, and neuroprotection. Small clinical trials and observational studies indicate that ALLO may have cognitive benefits, but no large-scale, definitive meta-analysis confirms a 20% improvement in AD patients. Mitochondrial dysfunction plays a vital role in the pathogenesis of numerous neurological diseases due to the high-energy demand and sensitivity of neurons to oxidative stress. Reduced mitochondrial function leads to amyloid-beta plaques and tau tangles accumulation in AD. In Parkinson's disease (PD), mitochondrial dysfunction resulting from the PINK1 or Parkin genes leads to energy deficiencies and the accumulation of toxic byproducts. Mutations in genes such as SOD1, C9orf72, and TDP-43 have been associated with mitochondrial dysfunction in amyotrophic lateral sclerosis (ALS). Moreover, studies on these neurodegenerative diseases suggest that inflammation is not merely a consequence of neurodegeneration but is also an essential factor in this process. Many neurological disorders involve multifaceted interactions between genetics, the environment, and immune responses, making it difficult to pinpoint their exact causes. Future research aims to overcome these hurdles through genetic advances, regenerative medicine, and personalized therapies. Cutting-edge technologies such as artificial intelligence and high-throughput screening are expected to accelerate drug discovery and improve diagnostic accuracy. Increasing collaboration between interdisciplinary fields such as bioinformatics, neuroscience, and immunology will lead to innovative treatment strategies. This comprehensive review discusses the therapeutic effects of enzymes, neurosteroids, and synthetic steroids in different neurodegenerative diseases, particularly AD, PD, ALS, and MS. Potential challenges in the therapeutic use of neurosteroids, such as the limited bioavailability and off-target effects of synthetic steroids, are also discussed, and an up-to-date and comprehensive review of the impact of these steroids on neurodegenerative disorders is presented.
Collapse
Affiliation(s)
- Refik Servi
- Fırat University, Faculty of Medicine, Department of Anatomy, Elazığ.
| | | | - Feyza Aksu
- Fırat University, Faculty of Medicine, Department of Anatomy, Elazığ.
| | - Süleyman Servi
- Fırat University, Faculty of Science, Department of Chemistry, Elazığ.
| |
Collapse
|
2
|
Bourque M, Morissette M, Isenbrandt A, Giatti S, Melcangi RC, Carta M, Frau R, Bortolato M, Soulet D, Di Paolo T. Effect of 5-alpha reductase inhibitors in animal models of Parkinson's disease. Front Neuroendocrinol 2024; 75:101156. [PMID: 39353534 DOI: 10.1016/j.yfrne.2024.101156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Parkinson's disease (PD) is characterized by motor symptoms due to loss of brain dopamine and non-motor symptoms, including gastrointestinal disorders. Although there is no cure for PD, symptomatic treatments are available. L-Dopa is the gold standard PD therapy, but most patients develop dyskinesias (LID), which are challenging to manage. Amantadine is recognized as the most effective drug for LID, but its adverse effects limit the use in patients. Here we review how 5α-reductase inhibitors (5ARIs), drugs used to treat benign prostatic hyperplasia and alopecia, exhibit beneficial effects in PD animal models. 5ARIs show neuroprotective properties in brain and gut dopaminergic systems, and reduce dyskinesias in rodent model of PD. Additionally, the 5ARI finasteride dampened dopaminergic-induced drug gambling in PD patients. Neuroprotection and antidyskinetic activities of 5ARIs in animal models of PD suggest their potential repurposing in men with PD to address gut dysfunction, protect brain DA and inhibit dyskinesias.
Collapse
Affiliation(s)
- Mélanie Bourque
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Marc Morissette
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Amandine Isenbrandt
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada
| | - Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Manolo Carta
- Department of Biomedical Sciences, "Guy Everett Laboratory", University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, "Guy Everett Laboratory", University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT 84112, USA
| | - Denis Soulet
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada
| | - Thérèse Di Paolo
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada.
| |
Collapse
|
3
|
King JD, Elliott T, Pitman A. Steroid-induced mania in a patient with previously well-controlled organic bipolar 1-like affective disorder secondary to acquired brain injury: case report and literature review. DISCOVER MENTAL HEALTH 2024; 4:8. [PMID: 38453827 PMCID: PMC10920485 DOI: 10.1007/s44192-024-00061-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
Steroid-induced neuropsychiatric sequelae are common, and pose significant risks to people usually receiving glucocorticoids in the context of physical illness. Steroid-induced mania and hypomania are the most common of the acute complications, yet despite great progress in understandings in neurophysiology there are no recent studies which review the factors which might predict who will experience this severe complication, nor are there consensus guidelines on management. We report the unusual case of a woman in her 50s admitted to a psychiatric unit with steroid-induced mania despite compliance with two mood stabilisers, several days after the administration of a Dexamethasone and Docetaxel chemotherapy regime adjunctive to lumpectomy for breast cancer. She had previously been diagnosed with an organic affective disorder (with classical bipolar 1 pattern) following severe ventriculitis related to ventricular drain insertion for obstructive hydrocephalus secondary to a colloid cyst. She had no psychiatric illness before this brain injury, but has a maternal history of idiopathic bipolar 1 affective disorder. Her episode of steroid-induced mania resolved following use of sedative medications, continuation of her existing mood stabilisers, and reductions of the steroid dosing in collaboration with her oncology team, which also protected her from further manic relapses during continued chemotherapy. Established mental illness, a family history, and acquired brain injury may reflect risk factors for steroid-induced mania through currently unclear pathways. Future epidemiological studies could better confirm these observations, and basic neuroscience may look to further explore the role of extrinsic glucocorticoids in the pathophysiology of affective disorders.
Collapse
Affiliation(s)
- Jacob D King
- Division of Psychiatry, Imperial College London, London, UK.
- Central and North West London NHS Foundation Trust, London, UK.
- Camden and Islington NHS Foundation Trust, London, UK.
| | - Thomas Elliott
- Camden and Islington NHS Foundation Trust, London, UK
- East London Foundation Trust, London, UK
| | - Alexandra Pitman
- Camden and Islington NHS Foundation Trust, London, UK
- Division of Psychiatry, University College London, London, UK
| |
Collapse
|
4
|
Ni RJ, Wang YY, Pu WJ, Wei YY, Wei JX, Zhao LS, Ma XH. Differential effects of sleep deprivation on behavior and microglia in a brain-region-specific manner in young and aged male mice. Brain Behav Immun 2024; 117:12-19. [PMID: 38157946 DOI: 10.1016/j.bbi.2023.12.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024] Open
Abstract
Microglia, resident immune cells in the central nervous system, constantly monitor the state of the surrounding brain activity. The animal model induced by sleep deprivation (SD) is widely used to study the pathophysiological mechanisms of insomnia and bipolar disorder. However, it remains unclear whether SD affects behaviors in young and aged male mice and microglia in various brain regions. In this study, we confirmed brain region-specific changes in microglial density and morphology in the accumbens nucleus (Acb), amygdala (AMY), cerebellum (Cb), corpus callosum (cc), caudate putamen, hippocampus (HIP), hypothalamus (HYP), medial prefrontal cortex (mPFC), and thalamus (TH) of young mice. In addition, the density of microglia in old mice was higher than that in young mice. Compared with young mice, old mice showed a markedly increased microglial size, decreased total length of microglial processes, and decreased maximum length. Importantly, we found that 48-h SD decreased microglial density and morphology in old mice, whereas SD increased microglial density and morphology in most observed brain regions in young mice. SD-induced hyperactivity was observed only in young mice but not in old mice. Moreover, microglial density (HIP, AMY, mPFC, CPu) was significantly positively correlated with behaviors in SD- and vehicle-treated young mice. Contrarily, negative correlations were shown between the microglial density (cc, Cb, TH, HYP, Acb, AMY) and behaviors in vehicle-treated young and old mice. These results suggest that SD dysregulates the homeostatic state of microglia in a region- and age-dependent manner. Microglia may be involved in regulating age-related behavioral responses to SD.
Collapse
Affiliation(s)
- Rong-Jun Ni
- Mental Health Center and Psychiatric Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, Sichuan 610044, China.
| | - Yi-Yan Wang
- Mental Health Center and Psychiatric Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, Sichuan 610044, China
| | - Wen-Jun Pu
- Mental Health Center and Psychiatric Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, Sichuan 610044, China
| | - Ying-Ying Wei
- Mental Health Center and Psychiatric Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, Sichuan 610044, China
| | - Jin-Xue Wei
- Mental Health Center and Psychiatric Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, Sichuan 610044, China
| | - Lian-Sheng Zhao
- Mental Health Center and Psychiatric Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, Sichuan 610044, China
| | - Xiao-Hong Ma
- Mental Health Center and Psychiatric Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, Sichuan 610044, China.
| |
Collapse
|
5
|
Que M, Li Y, Wang X, Zhan G, Luo X, Zhou Z. Role of astrocytes in sleep deprivation: accomplices, resisters, or bystanders? Front Cell Neurosci 2023; 17:1188306. [PMID: 37435045 PMCID: PMC10330732 DOI: 10.3389/fncel.2023.1188306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/05/2023] [Indexed: 07/13/2023] Open
Abstract
Sleep plays an essential role in all studied animals with a nervous system. However, sleep deprivation leads to various pathological changes and neurobehavioral problems. Astrocytes are the most abundant cells in the brain and are involved in various important functions, including neurotransmitter and ion homeostasis, synaptic and neuronal modulation, and blood-brain barrier maintenance; furthermore, they are associated with numerous neurodegenerative diseases, pain, and mood disorders. Moreover, astrocytes are increasingly being recognized as vital contributors to the regulation of sleep-wake cycles, both locally and in specific neural circuits. In this review, we begin by describing the role of astrocytes in regulating sleep and circadian rhythms, focusing on: (i) neuronal activity; (ii) metabolism; (iii) the glymphatic system; (iv) neuroinflammation; and (v) astrocyte-microglia cross-talk. Moreover, we review the role of astrocytes in sleep deprivation comorbidities and sleep deprivation-related brain disorders. Finally, we discuss potential interventions targeting astrocytes to prevent or treat sleep deprivation-related brain disorders. Pursuing these questions would pave the way for a deeper understanding of the cellular and neural mechanisms underlying sleep deprivation-comorbid brain disorders.
Collapse
Affiliation(s)
- Mengxin Que
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yujuan Li
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Wang
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Gaofeng Zhan
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiao Luo
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Zhou
- Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Department of Anesthesiology, Tongji Medical College, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Corsi S, Scheggi S, Pardu A, Braccagni G, Caruso D, Cioffi L, Diviccaro S, Gentile M, Fanni S, Stancampiano R, Gambarana C, Melcangi RC, Frau R, Carta M. Pregnenolone for the treatment of L-DOPA-induced dyskinesia in Parkinson's disease. Exp Neurol 2023; 363:114370. [PMID: 36878398 DOI: 10.1016/j.expneurol.2023.114370] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/02/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023]
Abstract
Growing preclinical and clinical evidence highlights neurosteroid pathway imbalances in Parkinson's Disease (PD) and L-DOPA-induced dyskinesias (LIDs). We recently reported that 5α-reductase (5AR) inhibitors dampen dyskinesias in parkinsonian rats; however, unraveling which specific neurosteroid mediates this effect is critical to optimize a targeted therapy. Among the 5AR-related neurosteroids, striatal pregnenolone has been shown to be increased in response to 5AR blockade and decreased after 6-OHDA lesions in the rat PD model. Moreover, this neurosteroid rescued psychotic-like phenotypes by exerting marked antidopaminergic activity. In light of this evidence, we investigated whether pregnenolone might dampen the appearance of LIDs in parkinsonian drug-naïve rats. We tested 3 escalating doses of pregnenolone (6, 18, 36 mg/kg) in 6-OHDA-lesioned male rats and compared the behavioral, neurochemical, and molecular outcomes with those induced by the 5AR inhibitor dutasteride, as positive control. The results showed that pregnenolone dose-dependently countered LIDs without affecting L-DOPA-induced motor improvements. Post-mortem analyses revealed that pregnenolone significantly prevented the increase of validated striatal markers of dyskinesias, such as phospho-Thr-34 DARPP-32 and phospho-ERK1/2, as well as D1-D3 receptor co-immunoprecipitation in a fashion similar to dutasteride. Moreover, the antidyskinetic effect of pregnenolone was paralleled by reduced striatal levels of BDNF, a well-established factor associated with the development of LIDs. In support of a direct pregnenolone effect, LC/MS-MS analyses revealed that striatal pregnenolone levels strikingly increased after the exogenous administration, with no significant alterations in downstream metabolites. All these data suggest pregnenolone as a key player in the antidyskinetic properties of 5AR inhibitors and highlight this neurosteroid as an interesting novel tool to target LIDs in PD.
Collapse
Affiliation(s)
- Sara Corsi
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy
| | - Simona Scheggi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, SI, Italy
| | - Alessandra Pardu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy
| | - Giulia Braccagni
- Department of Molecular and Developmental Medicine, University of Siena, Siena, SI, Italy
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Lucia Cioffi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Silvia Diviccaro
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Mauro Gentile
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy
| | - Silvia Fanni
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy; Basal Ganglia Pathophysiology Unit, Department Experimental Medical Science, Lund University, Sweden
| | | | - Carla Gambarana
- Department of Molecular and Developmental Medicine, University of Siena, Siena, SI, Italy
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy; "Guy Everett Laboratory", Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.
| | - Manolo Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy.
| |
Collapse
|
7
|
Sleep Deprivation Induces Dopamine System Maladaptation and Escalated Corticotrophin-Releasing Factor Signaling in Adolescent Mice. Mol Neurobiol 2023; 60:3190-3209. [PMID: 36813955 DOI: 10.1007/s12035-023-03258-2] [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: 06/17/2022] [Accepted: 02/07/2023] [Indexed: 02/24/2023]
Abstract
Sleep disruption is highly associated with the pathogenesis and progression of a wild range of psychiatric disorders. Furthermore, appreciable evidence shows that experimental sleep deprivation (SD) on humans and rodents evokes anomalies in the dopaminergic (DA) signaling, which are also implicated in the development of psychiatric illnesses such as schizophrenia or substance abuse. Since adolescence is a vital period for the maturation of the DA system as well as the occurrence of mental disorders, the present studies aimed to investigate the impacts of SD on the DA system of adolescent mice. We found that 72 h SD elicited a hyperdopaminergic status, with increased sensitivity to the novel environment and amphetamine (Amph) challenge. Also, altered neuronal activity and expression of striatal DA receptors were noticed in the SD mice. Moreover, 72 h SD influenced the immune status in the striatum, with reduced microglial phagocytic capacity, primed microglial activation, and neuroinflammation. The abnormal neuronal and microglial activity were putatively provoked by the enhanced corticotrophin-releasing factor (CRF) signaling and sensitivity during the SD period. Together, our findings demonstrated the consequences of SD in adolescents including aberrant neuroendocrine, DA system, and inflammatory status. Sleep insufficiency is a risk factor for the aberration and neuropathology of psychiatric disorders.
Collapse
|
8
|
Frau R, Melis M. Sex-specific susceptibility to psychotic-like states provoked by prenatal THC exposure: Reversal by pregnenolone. J Neuroendocrinol 2023; 35:e13240. [PMID: 36810840 DOI: 10.1111/jne.13240] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/23/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
Sociocultural attitudes towards cannabis legalization contribute to the common misconception that it is a relatively safe drug and its use during pregnancy poses no risk to the fetus. However, longitudinal studies demonstrate that maternal cannabis exposure results in adverse outcomes in the offspring, with a heightened risk for developing psychopathology. One of the most reported psychiatric outcomes is the proneness to psychotic-like experiences during childhood. How exposure to cannabis during gestation increases psychosis susceptibility in children and adolescents remains elusive. Preclinical research has indicated that in utero exposure to the major psychoactive component of cannabis, delta-9-tetrahydrocannabinol (THC), deranges brain developmental trajectories towards vulnerable psychotic-like endophenotypes later in life. Here, we present how prenatal THC exposure (PCE) deregulates mesolimbic dopamine development predisposing the offspring to schizophrenia-relevant phenotypes, exclusively when exposed to environmental challenges, such as stress or THC. Detrimental effects of PCE are sex-specific because female offspring do not display psychotic-like outcomes upon exposure to these challenges. Moreover, we present how pregnenolone, a neurosteroid that showed beneficial properties on the effects elicited by cannabis intoxication, normalizes mesolimbic dopamine function and rescues psychotic-like phenotypes. We, therefore, suggest this neurosteroid as a safe "disease-modifying" aid to prevent the onset of psychoses in vulnerable individuals. Our findings corroborate clinical evidence and highlight the relevance of early diagnostic screening and preventative strategies for young individuals at risk for mental diseases, such as male PCE offspring.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
- The Guy Everett Laboratory for Neuroscience, University of Cagliari, Cagliari, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
| |
Collapse
|
9
|
Acute stress impairs sensorimotor gating via the neurosteroid allopregnanolone in the prefrontal cortex. Neurobiol Stress 2022; 21:100489. [DOI: 10.1016/j.ynstr.2022.100489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/07/2022] [Accepted: 09/20/2022] [Indexed: 11/21/2022] Open
|
10
|
Margiani G, Castelli MP, Pintori N, Frau R, Ennas MG, Pagano Zottola AC, Orrù V, Serra V, Fiorillo E, Fadda P, Marsicano G, De Luca MA. Adolescent self-administration of the synthetic cannabinoid receptor agonist JWH-018 induces neurobiological and behavioral alterations in adult male mice. Psychopharmacology (Berl) 2022; 239:3083-3102. [PMID: 35943523 PMCID: PMC9481487 DOI: 10.1007/s00213-022-06191-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 07/10/2022] [Indexed: 12/02/2022]
Abstract
RATIONALE The use of synthetic cannabinoid receptor agonists (SCRAs) is growing among adolescents, posing major medical and psychiatric risks. JWH-018 represents the reference compound of SCRA-containing products. OBJECTIVES This study was performed to evaluate the enduring consequences of adolescent voluntary consumption of JWH-018. METHODS The reinforcing properties of JWH-018 were characterized in male CD1 adolescent mice by intravenous self-administration (IVSA). Afterwards, behavioral, neurochemical, and molecular evaluations were performed at adulthood. RESULTS Adolescent mice acquired operant behavior (lever pressing, Fixed Ratio 1-3; 7.5 µg/kg/inf); this behavior was specifically directed at obtaining JWH-018 since it increased under Progressive Ratio schedule of reinforcement, and was absent in vehicle mice. JWH-018 IVSA was reduced by pretreatment of the CB1-antagonist/inverse agonist AM251. Adolescent exposure to JWH-018 by IVSA increased, at adulthood, both nestlet shredding and marble burying phenotypes, suggesting long-lasting repetitive/compulsive-like behavioral effects. JWH-018 did not affect risk proclivity in the wire-beam bridge task. In adult brains, there was an increase of ionized calcium binding adaptor molecule 1 (IBA-1) positive cells in the caudate-putamen (CPu) and nucleus accumbens (NAc), along with a decrease of glial fibrillary acidic protein (GFAP) immunoreactivity in the CPu. These glial alterations in adult brains were coupled with an increase of the chemokine RANTES and a decrease of the cytokines IL2 and IL13 in the cortex, and an increase of the chemokine MPC1 in the striatum. CONCLUSIONS This study suggests for the first time that male mice self-administer the prototypical SCRA JWH-018 during adolescence. The adolescent voluntary consumption of JWH-018 leads to long-lasting behavioral and neurochemical aberrations along with glia-mediated inflammatory responses in adult brains.
Collapse
Affiliation(s)
- Giulia Margiani
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | | | - Nicholas Pintori
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.,"Guy Everett" Laboratory, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Maria Grazia Ennas
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Antonio C Pagano Zottola
- INSERM, U1215 NeuroCentre Magendie, Bordeaux, France.,University of Bordeaux, Bordeaux, France.,Institut de Biochimie et Génétique Cellulaires, UMR 5095, Bordeaux, France
| | - Valeria Orrù
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Lanusei, Italy
| | - Valentina Serra
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Lanusei, Italy
| | - Edoardo Fiorillo
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Lanusei, Italy
| | - Paola Fadda
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.,Institute of Neuroscience-Cagliari, National Research Council (CNR), Cagliari, Italy
| | - Giovanni Marsicano
- INSERM, U1215 NeuroCentre Magendie, Bordeaux, France.,University of Bordeaux, Bordeaux, France
| | | |
Collapse
|
11
|
Chen S, Xie Y, Li Y, Fan X, Xing F, Mao Y, Xing N, Wang J, Yang J, Wang Z, Yuan J. Sleep deprivation and recovery sleep affect healthy male resident’s pain sensitivity and oxidative stress markers: The medial prefrontal cortex may play a role in sleep deprivation model. Front Mol Neurosci 2022; 15:937468. [PMID: 36061364 PMCID: PMC9434020 DOI: 10.3389/fnmol.2022.937468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Sleep is essential for the body’s repair and recovery, including supplementation with antioxidants to maintain the balance of the body’s redox state. Changes in sleep patterns have been reported to alter this repair function, leading to changes in disease susceptibility or behavior. Here, we recruited healthy male physicians and measured the extent of the effect of overnight sleep deprivation (SD) and recovery sleep (RS) on nociceptive thresholds and systemic (plasma-derived) redox metabolism, namely, the major antioxidants glutathione (GSH), catalase (CAT), malondialdehyde (MDA), and superoxide dismutase (SOD). Twenty subjects underwent morning measurements before and after overnight total SD and RS. We found that one night of SD can lead to increased nociceptive hypersensitivity and the pain scores of the Numerical Rating Scale (NRS) and that one night of RS can reverse this change. Pre- and post-SD biochemical assays showed an increase in MDA levels and CAT activity and a decrease in GSH levels and SOD activity after overnight SD. Biochemical assays before and after RS showed a partial recovery of MDA levels and a basic recovery of CAT activity to baseline levels. An animal study showed that SD can cause a significant decrease in the paw withdrawal threshold and paw withdrawal latency in rats, and after 4 days of unrestricted sleep, pain thresholds can be restored to normal. We performed proteomics in the rat medial prefrontal cortex (mPFC) and showed that 37 proteins were significantly altered after 6 days of SD. Current findings showed that SD causes nociceptive hyperalgesia and oxidative stress, and RS can restore pain thresholds and repair oxidative stress damage in the body. However, one night of RS is not enough for repairing oxidative stress damage in the human body.
Collapse
Affiliation(s)
- Shuhan Chen
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, China
| | - Yanle Xie
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, China
| | - Yize Li
- Department of Anesthesiology, Tianjin Research Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaochong Fan
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, China
| | - Fei Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, China
| | - Yuanyuan Mao
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, China
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, China
| | - Jingping Wang
- Massachusetts General Hospital Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, United States
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, China
| | - Zhongyu Wang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, China
- Zhongyu Wang,
| | - Jingjing Yuan
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, China
- *Correspondence: Jingjing Yuan,
| |
Collapse
|
12
|
Floris G, Scheggi S, Pes R, Bortolato M. The steroidogenic inhibitor finasteride reverses pramipexole-induced alterations in probability discounting. Brain Res Bull 2022; 181:157-166. [PMID: 35122898 PMCID: PMC9012661 DOI: 10.1016/j.brainresbull.2022.01.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/21/2022] [Accepted: 01/29/2022] [Indexed: 12/28/2022]
Abstract
Pramipexole is a potent agonist of D3 and D2 dopamine receptors, currently approved for clinical use in Parkinson's disease (PD) and restless leg syndrome. Several studies have shown that pramipexole significantly increases the risk of pathological gambling and impulse-control disorders. While these iatrogenic complications can impose a severe social and financial burden, their treatment poses serious clinical challenges. Our group previously reported that the steroidogenic inhibitor finasteride reduced pathological gambling severity in PD patients who developed this complication following pramipexole treatment. To study the mechanisms underlying these effects, here we tested the impact of finasteride in a rat model of pramipexole-induced alterations of probability discounting. We previously showed that, in rats exposed to low doses of the monoamine-depleting agent reserpine (1mg/kg/day, SC), pramipexole (0.3mg/kg/day, SC) increased the propensity to engage in disadvantageous choices. This effect was paralleled by a marked D3 receptor upregulation in the nucleus accumbens. First, we tested how finasteride (25-50mg/kg, IP) intrinsically affects probability discounting. While the highest dose of finasteride produced a marked lack of interest in lever pressing (manifested as a significant increase in omissions), the 25mg/kg (IP) dose did not intrinsically modify probability discounting. However, this finasteride regimen significantly reduced the adverse effects of reserpine and pramipexole in probability discounting by diminishing rats' propensity to engage in highly disadvantageous probabilistic choices. The same regimen also reversed the upregulation of D3 receptors in the nucleus accumbens induced by reserpine and pramipexole. These findings confirm that finasteride opposes the impulsivity caused by pramipexole and suggest that this effect may be underpinned by a normalizing effect on D3 receptor expression in the nucleus accumbens.
Collapse
Affiliation(s)
- Gabriele Floris
- Dept. of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City (UT), USA.
| | - Simona Scheggi
- Dept. of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City (UT), USA; Dept. of Molecular and Developmental Medicine, School of Medicine, University of Siena, ITALY
| | - Romina Pes
- Dept. of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence (KS), USA
| | - Marco Bortolato
- Dept. of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City (UT), USA; Dept. of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence (KS), USA.
| |
Collapse
|
13
|
Bortolato M, Coffey BJ, Gabbay V, Scheggi S. Allopregnanolone: The missing link to explain the effects of stress on tic exacerbation? J Neuroendocrinol 2022; 34:e13022. [PMID: 34423500 PMCID: PMC8800948 DOI: 10.1111/jne.13022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/19/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022]
Abstract
The neurosteroid allopregnanolone (3α-hydroxy-5α-pregnan-20-one; AP) elicits pleiotropic effects in the central nervous system, ranging from neuroprotective and anti-inflammatory functions to the regulation of mood and emotional responses. Several lines of research show that the brain rapidly produces AP in response to acute stress to reduce the allostatic load and enhance coping. These effects not only are likely mediated by GABAA receptor activation but also result from the contributions of other mechanisms, such as the stimulation of membrane progesterone receptors. In keeping with this evidence, AP has been shown to exert rapid, potent antidepressant properties and has been recently approved for the therapy of moderate-to-severe postpartum depression. In addition to depression, emerging evidence points to the potential of AP as a therapy for other neuropsychiatric disorders, including anxiety, seizures, post-traumatic stress disorder and cognitive problems. Although this evidence has spurred interest in further therapeutic applications of AP, some investigations suggest that this neurosteroid may also be associated with adverse events in specific disorders. For example, our group has recently documented that AP increases tic-like manifestations in several animal models of tic disorders; furthermore, our results indicate that inhibiting AP synthesis and signalling reduces the exacerbation of tic severity associated with acute stress. Although the specific mechanisms of these effects remain partially elusive, our findings point to the possibility that the GABAergic activation by AP may also lead to disinhibitory effects, which could interfere with the ability of patients to suppress their tics. Future studies will be necessary to verify whether these mechanisms may apply to other externalising manifestations, such as impulse-control problems and manic symptoms.
Collapse
Affiliation(s)
- Marco Bortolato
- Department of Pharmacology and ToxicologyCollege of PharmacyUniversity of UtahSalt Lake CityUTUSA
- Research Consortium on NeuroEndocrine Causes of Tics (ReConNECT)
| | - Barbara J. Coffey
- Research Consortium on NeuroEndocrine Causes of Tics (ReConNECT)
- Department of Psychiatry and Behavioral ScienceMiller School of MedicineUniversity of MiamiMiamiFLUSA
| | - Vilma Gabbay
- Research Consortium on NeuroEndocrine Causes of Tics (ReConNECT)
- Department of Psychiatry and Behavioral SciencesAlbert Einstein College of MedicineBronxNYUSA
| | - Simona Scheggi
- Department of Molecular and Developmental MedicineSchool of MedicineUniversity of SienaSienaItaly
| |
Collapse
|
14
|
Arora S, Dharavath RN, Bansal Y, Bishnoi M, Kondepudi KK, Chopra K. Neurobehavioral alterations in a mouse model of chronic partial sleep deprivation. Metab Brain Dis 2021; 36:1315-1330. [PMID: 33740181 DOI: 10.1007/s11011-021-00693-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 02/12/2021] [Indexed: 12/23/2022]
Abstract
The night shift paradigm induces a state of chronic partial sleep deprivation (CPSD) and enhances the vulnerability to neuronal dysfunction. However, the specific neuronal impact of CPSD has not been thoroughly explored to date. In the current study, the night shift condition was mimicked in female Swiss albino mice. The classical sleep deprivation model, i.e., Modified Multiple Platform (MMP) method, was used for 8 h/day from Monday to Friday with Saturday and Sunday as a weekend off for nine weeks. Following nine weeks of night shift schedule, their neurobehavioral profile and physiological parameters were assessed along with the activity of the mitochondrial complexes, oxidative stress, serotonin levels, and inflammatory markers in the brain. Mice showed an overall hyperactive behavioral profile including hyperlocomotion, aggression, and stereotyped behavior accompanied by decreased activity of mitochondrial enzymes and serotonin levels, increased oxidative stress and inflammatory markers in whole brain homogenates. Collectively, the study points towards the occurrence of a hyperactive behavioral profile akin to mania and psychosis as a potential consequence of CPSD.
Collapse
Affiliation(s)
- Shiyana Arora
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India
| | - Ravinder Naik Dharavath
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India
| | - Yashika Bansal
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India
| | - Mahendra Bishnoi
- Food and Nutritional Biotechnology Laboratory, National Agri-Food Biotechnology Institute, SAS Nagar, Punjab, 140306, India
| | - Kanthi Kiran Kondepudi
- Food and Nutritional Biotechnology Laboratory, National Agri-Food Biotechnology Institute, SAS Nagar, Punjab, 140306, India
| | - Kanwaljit Chopra
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Sector 14, 160014, Chandigarh, India.
| |
Collapse
|
15
|
Sagheddu C, Traccis F, Serra V, Congiu M, Frau R, Cheer JF, Melis M. Mesolimbic dopamine dysregulation as a signature of information processing deficits imposed by prenatal THC exposure. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110128. [PMID: 33031862 DOI: 10.1016/j.pnpbp.2020.110128] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/02/2020] [Accepted: 10/03/2020] [Indexed: 11/17/2022]
Abstract
Cannabis is the illicit drug most widely used by pregnant women worldwide. Its growing acceptance and legalization have markedly increased the risks of child psychopathology, including psychotic-like experiences, which lowers the age of onset for a first psychotic episode. As the majority of patients with schizophrenia go through a premorbid condition long before this occurs, understanding neurobiological underpinnings of the prodromal stage of the disease is critical to improving illness trajectories and therapeutic outcomes. We have previously shown that male rat offspring prenatally exposed to Δ9-tetrahydrocannabinol (THC), a rat model of prenatal cannabinoid exposure (PCE), exhibit extensive molecular and synaptic changes in dopaminergic neurons of the ventral tegmental area (VTA), converging on a hyperdopaminergic state. This leads to a silent psychotic-like endophenotype that is unmasked by a single exposure to THC. Here, we further characterized the VTA dopamine neuron and sensorimotor gating functions of PCE rats exposed to acute stress or a challenge of the D2 receptor agonist apomorphine, by using in vivo single-unit extracellular recordings and Prepulse Inhibition (PPI) analyses. At pre-puberty, PCE male rat offspring display a reduced population activity of VTA dopamine neurons in vivo, the majority of which are tonically active. PCE male progeny also exhibit enhanced sensitivity to dopamine D2 (DAD2) receptor activation and a vulnerability to acute stress, which is associated with compromised sensorimotor gating functions. This data extends our knowledge of the multifaceted sequelae imposed by PCE in the mesolimbic dopamine system of male pre-adolescent rats, which renders a neural substrate highly susceptible to subsequent challenges that may trigger psychotic-like outcomes.
Collapse
Affiliation(s)
- Claudia Sagheddu
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
| | - Francesco Traccis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
| | - Valeria Serra
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
| | - Mauro Congiu
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
| | - Joseph F Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Miriam Melis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy.
| |
Collapse
|
16
|
Orrù M, Strathman HJ, Floris G, Scheggi S, Levant B, Bortolato M. The adverse effects of pramipexole on probability discounting are not reversed by acute D 2 or D 3 receptor antagonism. Eur Neuropsychopharmacol 2020; 32:104-119. [PMID: 31983530 PMCID: PMC9325630 DOI: 10.1016/j.euroneuro.2020.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/06/2019] [Accepted: 01/07/2020] [Indexed: 02/08/2023]
Abstract
Pramipexole (PPX) is a D2 and D3 dopamine receptor agonist approved for clinical use, which is associated with a higher risk of impulse-control disorders. Using a rat model, we recently found that low doses of the monoamine-depleting agent reserpine (RES; 1 mg/kg/day, SC) dramatically increased the untoward effects of PPX (0.3 mg/kg/day, SC) on probability discounting, a key impulsivity function. To further understand the neurobehavioral mechanisms underlying these effects, we first tested whether the combination of PPX and RES may lead to a generalized enhancement in risk taking, as tested in the suspended wire-beam paradigm. The association of RES and PPX did not augment the proclivity of rats to cross the bridge in order to obtain a reward, suggesting that the effects of RES and PPX on probability discounting do not reflect a generalized increase in impulsivity. We then studied what receptors mediate the effects of PPX in RES-treated rats. The combination of RES and PPX increased membrane expression and binding of D3, but not D2 dopamine receptors, in the nucleus accumbens. However, the behavioral effects of PPX and RES were not reduced by acute treatments with the D2/D3 receptor antagonist raclopride (0.01-0.05 mg/kg, SC), the highly selective D2 receptor antagonist L-741,626 (0.1-1 mg/kg, SC) or the D3 receptor antagonists GR 103691 (0.1-0.3 mg/kg, SC) and SB 277011A (1-10 mg/kg, SC). These findings collectively suggest that the effects of PPX in probability discounting do not reflect generalized enhancements in impulsivity or acute dopamine D2 or D3 receptor activation.
Collapse
Affiliation(s)
- Marco Orrù
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Skaggs Hall, Room 3916, 30 S 2000 E, Salt Lake City, UT, Unites States
| | - Hunter J Strathman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Skaggs Hall, Room 3916, 30 S 2000 E, Salt Lake City, UT, Unites States
| | - Gabriele Floris
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Skaggs Hall, Room 3916, 30 S 2000 E, Salt Lake City, UT, Unites States
| | - Simona Scheggi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Skaggs Hall, Room 3916, 30 S 2000 E, Salt Lake City, UT, Unites States; Department of Molecular and Developmental Medicine, School of Medicine, University of Siena, Italy
| | - Beth Levant
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS, Unites States
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Skaggs Hall, Room 3916, 30 S 2000 E, Salt Lake City, UT, Unites States.
| |
Collapse
|
17
|
A Pilot Study on Sleep Quality, Forgiveness, Religion, Spirituality, and General Health of Women Living in a Homeless Mission. Holist Nurs Pract 2020; 34:49-56. [DOI: 10.1097/hnp.0000000000000362] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
18
|
Frau R, Traccis F, Bortolato M. Neurobehavioural complications of sleep deprivation: Shedding light on the emerging role of neuroactive steroids. J Neuroendocrinol 2020; 32:e12792. [PMID: 31505075 PMCID: PMC6982588 DOI: 10.1111/jne.12792] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/06/2019] [Accepted: 09/05/2019] [Indexed: 01/05/2023]
Abstract
Sleep deprivation (SD) is associated with a broad spectrum of cognitive and behavioural complications, including emotional lability and enhanced stress reactivity, as well as deficits in executive functions, decision making and impulse control. These impairments, which have profound negative consequences on the health and productivity of many individuals, reflect alterations of the prefrontal cortex (PFC) and its connectivity with subcortical regions. However, the molecular underpinnings of these alterations remain elusive. Our group and others have begun examining how the neurobehavioural outcomes of SD may be influenced by neuroactive steroids, a family of molecules deeply implicated in sleep regulation and the stress response. These studies have revealed that, similar to other stressors, acute SD leads to increased synthesis of the neurosteroid allopregnanolone in the PFC. Whereas this up-regulation is likely aimed at counterbalancing the detrimental impact of oxidative stress induced by SD, the increase in prefrontal allopregnanolone levels contributes to deficits in sensorimotor gating and impulse control, signalling a functional impairment of PFC. This scenario suggests that the synthesis of neuroactive steroids during acute SD may be enacted as a neuroprotective response in the PFC; however, such compensation may in turn set off neurobehavioural complications by interfering with the corticolimbic connections responsible for executive functions and emotional regulation.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato (CA), Italy
- National Institute of Neuroscience (INN), University of Cagliari, Monserrato (CA), Italy
| | - Francesco Traccis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato (CA), Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City (UT), USA
| |
Collapse
|
19
|
Abstract
Inhibitors of 5α-steroid reductase are drugs used to treat androgen-dependent conditions including prostate diseases and androgenic alopecia. Finasteride was the first on the market and is currently the most widely used inhibitor. Dutasteride was the second inhibitor to be approved and has a similar safety profile. Common adverse events of treatment consist of sexual disorders and a negative affect balance. It was described that the prolonged use of 5α-steroid reductase inhibitors in patients with alopecia can cause persistent side effects called a post-finasteride syndrome (PFS), that is not just a simple coexistence of events, but rather a definite syndrome with an iatrogenic background. PFS occurs in susceptible individuals even after small doses of the drug and can last for a long time after the discontinuation of treatment. A deterioration in the quality of life in affected individuals does not justify use of the drug. Wider recognition of PFS symptoms, its incidence, course, prevention, and treatment possibilities will allow the indications for drug use to be reconsidered and treatment to be more personalized. Knowledge about PFS will also help to provide the best treatment for affected individuals and to properly educate patients before obtaining an informed consent for therapy with 5α-steroid reductase inhibitors.
Collapse
Affiliation(s)
- Radosław B Maksym
- The Department of Reproductive Health, Centre of Postgraduate Medical Education , Warsaw , Poland
| | - Anna Kajdy
- The Department of Reproductive Health, Centre of Postgraduate Medical Education , Warsaw , Poland
| | - Michał Rabijewski
- The Department of Reproductive Health, Centre of Postgraduate Medical Education , Warsaw , Poland
| |
Collapse
|
20
|
Godar SC, Cadeddu R, Floris G, Mosher LJ, Mi Z, Jarmolowicz DP, Scheggi S, Walf AA, Koonce CJ, Frye CA, Muma NA, Bortolato M. The Steroidogenesis Inhibitor Finasteride Reduces the Response to Both Stressful and Rewarding Stimuli. Biomolecules 2019; 9:biom9110749. [PMID: 31752360 PMCID: PMC6920809 DOI: 10.3390/biom9110749] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/14/2019] [Accepted: 11/18/2019] [Indexed: 01/15/2023] Open
Abstract
Finasteride (FIN) is the prototypical inhibitor of steroid 5α-reductase (5αR), the enzyme that catalyzes the rate-limiting step of the conversion of progesterone and testosterone into their main neuroactive metabolites. FIN is clinically approved for the treatment of benign prostatic hyperplasia and male baldness; while often well-tolerated, FIN has also been shown to cause or exacerbate psychological problems in vulnerable subjects. Evidence on the psychological effects of FIN, however, remains controversial, in view of inconsistent clinical reports. Here, we tested the effects of FIN in a battery of tests aimed at capturing complementary aspects of mood regulation and stress reactivity in rats. FIN reduced exploratory, incentive, prosocial, and risk-taking behavior; furthermore, it decreased stress coping, as revealed by increased immobility in the forced-swim test (FST). This last effect was also observed in female and orchiectomized male rats, suggesting that the mechanism of action of FIN does not primarily reflect changes in gonadal steroids. The effects of FIN on FST responses were associated with a dramatic decrease in corticotropin release hormone (CRH) mRNA and adrenocorticotropic hormone (ACTH) levels. These results suggest that FIN impairs stress reactivity and reduces behavioral activation and impulsive behavior by altering the function of the hypothalamus-pituitary-adrenal (HPA) axis.
Collapse
Affiliation(s)
- Sean C. Godar
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake, UT 84112, USA; (S.C.G.); (R.C.); (G.F.); (L.J.M.); (S.S.)
| | - Roberto Cadeddu
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake, UT 84112, USA; (S.C.G.); (R.C.); (G.F.); (L.J.M.); (S.S.)
| | - Gabriele Floris
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake, UT 84112, USA; (S.C.G.); (R.C.); (G.F.); (L.J.M.); (S.S.)
| | - Laura J. Mosher
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake, UT 84112, USA; (S.C.G.); (R.C.); (G.F.); (L.J.M.); (S.S.)
- Department of Pharmacology and Toxicology, School of Pharmacy; Lawrence, KS 66045, USA; (Z.M.); (N.A.M.)
| | - Zhen Mi
- Department of Pharmacology and Toxicology, School of Pharmacy; Lawrence, KS 66045, USA; (Z.M.); (N.A.M.)
| | - David P. Jarmolowicz
- Department of Applied Behavioral Science; University of Kansas, Lawrence, KS 66045, USA;
- Cofrin Logan Center for Addiction Research and Treatment; University of Kansas, Lawrence, KS 66045, USA
| | - Simona Scheggi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake, UT 84112, USA; (S.C.G.); (R.C.); (G.F.); (L.J.M.); (S.S.)
| | - Alicia A. Walf
- Department of Cognitive Science, Rensselaer Polytechnic Institute, Troy, NY 12180, USA;
- Department of Psychology; The University at Albany-SUNY, Albany, NY 12222, USA; (C.J.K.); (C.A.F.)
| | - Carolyn J. Koonce
- Department of Psychology; The University at Albany-SUNY, Albany, NY 12222, USA; (C.J.K.); (C.A.F.)
| | - Cheryl A. Frye
- Department of Psychology; The University at Albany-SUNY, Albany, NY 12222, USA; (C.J.K.); (C.A.F.)
- Department of Biological Sciences; The University at Albany-SUNY, Albany, NY 12222, USA
- Center for Neuroscience, The University at Albany-SUNY, Albany, NY 12222, USA
- Comprehensive Neuropsychological Services, Albany, NY 12203, USA
| | - Nancy A. Muma
- Department of Pharmacology and Toxicology, School of Pharmacy; Lawrence, KS 66045, USA; (Z.M.); (N.A.M.)
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake, UT 84112, USA; (S.C.G.); (R.C.); (G.F.); (L.J.M.); (S.S.)
- Correspondence:
| |
Collapse
|
21
|
Frau R, Miczán V, Traccis F, Aroni S, Pongor CI, Saba P, Serra V, Sagheddu C, Fanni S, Congiu M, Devoto P, Cheer JF, Katona I, Melis M. Prenatal THC exposure produces a hyperdopaminergic phenotype rescued by pregnenolone. Nat Neurosci 2019; 22:1975-1985. [PMID: 31611707 PMCID: PMC6884689 DOI: 10.1038/s41593-019-0512-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 09/11/2019] [Indexed: 12/21/2022]
Abstract
Increased legal availability of cannabis has led to a common misconception that it is a safe natural remedy for, amongst others, pregnancy-related ailments like morning sickness. Emerging clinical evidence, however, indicates that prenatal cannabis exposure (PCE) predisposes offspring to various neuropsychiatric disorders linked to aberrant dopaminergic function. Yet, our knowledge of how cannabis exposure affects the maturation of this neuromodulatory system remains limited. Here, we show that male, but not female, offspring of Δ9-tetrahydrocannabinol (THC)-exposed dams, a rat PCE model, exhibit extensive molecular and synaptic changes in dopaminergic neurons of the ventral tegmental area, including altered excitatory-to-inhibitory balance and switched polarity of long-term synaptic plasticity. The resulting hyperdopaminergic state leads to increased behavioral sensitivity to acute THC during pre-adolescence. The FDA-approved neurosteroid pregnenolone rescues synaptic defects and normalizes dopaminergic activity and behavior in PCE offspring, suggesting a therapeutic approach for offspring exposed to cannabis during pregnancy.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Vivien Miczán
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.,Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Francesco Traccis
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Sonia Aroni
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, Italy.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Csaba I Pongor
- Nikon Center of Excellence for Neuronal Imaging, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Pierluigi Saba
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Valeria Serra
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Claudia Sagheddu
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Silvia Fanni
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Mauro Congiu
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Paola Devoto
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Joseph F Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - István Katona
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, Italy.
| |
Collapse
|
22
|
Wendler E, de Souza CP, Dornellas APS, Santos LE, Ferreira ST, Galduróz JCF, Wöhr M, Schwarting RKW, Andreatini R. Mania-like elevated mood in rats: Enhanced 50-kHz ultrasonic vocalizations after sleep deprivation. Prog Neuropsychopharmacol Biol Psychiatry 2019; 88:142-150. [PMID: 29981775 DOI: 10.1016/j.pnpbp.2018.07.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 06/21/2018] [Accepted: 07/04/2018] [Indexed: 12/14/2022]
Abstract
Mania is characterized by elevated drive and mood but animal models of mania have often neglected elevated mood. Ultrasonic vocalizations (USV) of 50-kHz emitted by rats are thought to index the subject's positive affective state. Fifty-kHz USV emission is increased by amphetamine, an effect blocked by lithium administration. Sleep deprivation (SD) is an environmental model of mania and the present study evaluated SD effects on behavioral activity and USV emission, together with the impact of lithium treatment. Adult rats were submitted to 24h or 72h SD, and locomotor activity and USV emission were assessed. To test their sensitivity to a standard antimanic drug, these behavioral parameters were also evaluated after acute administration of lithium carbonate (25, 50 or 100 mg/kg, i.p.). Striatal monoamine content was measured post-mortem. SD (24h and 72h) led to increased locomotor activity, rearing behavior and 50-kHz USV emission, together with a change in the call profile characterized by an increase in the percentage of frequency-modulated 50-kHz USV, which may indicate the mania-like consequences of SD. Importantly, all SD effects were reverted by lithium administration. SD also led to a decrease in dopamine content in the ventral striatum, while increasing dopamine turnover. In conclusion, SD increased 50-kHz USV emission, an effect prevented by acute lithium administration. This suggests 50-kHz USV as a new marker for mania-like elevated mood, which shows construct validity (associated with increased dopaminergic tone), face validity (reflecting increased positive affect) and predictive validity (high sensitivity to lithium treatment).
Collapse
Affiliation(s)
- Etieli Wendler
- Department of Pharmacology, Biological Sciences Sector, Federal University of Paraná, Centro Politécnico, C.P. 19031, 81540-990 Curitiba, PR, Brazil
| | - Camila Pasquini de Souza
- Department of Pharmacology, Biological Sciences Sector, Federal University of Paraná, Centro Politécnico, C.P. 19031, 81540-990 Curitiba, PR, Brazil
| | - Ana Paula Segantine Dornellas
- Department of Pharmacology, Biological Sciences Sector, Federal University of Paraná, Centro Politécnico, C.P. 19031, 81540-990 Curitiba, PR, Brazil
| | - Luis Eduardo Santos
- Institute of Biophysics Carlos Chagas Filho & Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| | - Sergio T Ferreira
- Institute of Biophysics Carlos Chagas Filho & Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| | - José Carlos Fernandes Galduróz
- Department of Psychobiology, Universidade Federal de São Paulo, Rua Napoleão de Barros, 925, São Paulo, SP 04024-002, Brazil
| | - Markus Wöhr
- Behavioural Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Gutenbergstr. 18, 35032 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), Philipps-University of Marburg, Hans-Meerwein-Str. 6, 35032 Marburg, Germany
| | - Rainer K W Schwarting
- Behavioural Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Gutenbergstr. 18, 35032 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), Philipps-University of Marburg, Hans-Meerwein-Str. 6, 35032 Marburg, Germany
| | - Roberto Andreatini
- Department of Pharmacology, Biological Sciences Sector, Federal University of Paraná, Centro Politécnico, C.P. 19031, 81540-990 Curitiba, PR, Brazil.
| |
Collapse
|
23
|
Frau R, Bortolato M. Repurposing steroidogenesis inhibitors for the therapy of neuropsychiatric disorders: Promises and caveats. Neuropharmacology 2018; 147:55-65. [PMID: 29907425 DOI: 10.1016/j.neuropharm.2018.05.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/07/2018] [Accepted: 05/10/2018] [Indexed: 12/29/2022]
Abstract
Steroids exert a profound influence on behavioral reactivity, by modulating the functions of most neurotransmitters and shaping the impact of stress and sex-related variables on neural processes. This background - as well as the observation that most neuroactive steroids (including sex hormones, glucocorticoids and neurosteroids) are synthetized and metabolized by overlapping enzymatic machineries - points to steroidogenic pathways as a powerful source of targets for neuropsychiatric disorders. Inhibitors of steroidogenic enzymes have been developed and approved for a broad range of genitourinary and endocrine dysfunctions, opening to new opportunities to repurpose these drugs for the treatment of mental problems. In line with this idea, preliminary clinical and preclinical results from our group have shown that inhibitors of key steroidogenic enzymes, such as 5α-reductase and 17,20 desmolase-lyase, may have therapeutic efficacy in specific behavioral disorders associated with dopaminergic hyperfunction. While the lack of specificity of these effects raises potential concerns about endocrine adverse events, these initial findings suggest that steroidogenesis modulators with greater brain specificity may hold significant potential for the development of alternative therapies for psychiatric problems. This article is part of the Special Issue entitled 'Drug Repurposing: old molecules, new ways to fast track drug discovery and development for CNS disorders'.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato CA, Italy; Tourette Syndrome Center, University of Cagliari, Monserrato CA, Italy; Sleep Medicine Center, University of Cagliari, Monserrato CA, Italy; National Institute of Neuroscience (INN), University of Cagliari, Monserrato CA, Italy.
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
24
|
Mosher LJ, Godar SC, Morissette M, McFarlin KM, Scheggi S, Gambarana C, Fowler SC, Di Paolo T, Bortolato M. Steroid 5α-reductase 2 deficiency leads to reduced dominance-related and impulse-control behaviors. Psychoneuroendocrinology 2018; 91:95-104. [PMID: 29544191 PMCID: PMC5901899 DOI: 10.1016/j.psyneuen.2018.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/09/2018] [Accepted: 02/13/2018] [Indexed: 01/10/2023]
Abstract
The enzyme steroid 5α-reductase 2 (5αR2) catalyzes the conversion of testosterone into the potent androgen 5α-dihydrotestosterone. Previous investigations showed that 5αR2 is expressed in key brain areas for emotional and socio-affective reactivity, yet the role of this enzyme in behavioral regulation remains mostly unknown. Here, we profiled the behavioral characteristics of 5αR2 heterozygous (HZ) and knockout (KO) mice, as compared with their wild-type (WT) littermates. While male 5αR2 KO mice displayed no overt alterations in motoric, sensory, information-processing and anxiety-related behaviors, they exhibited deficits in neurobehavioral correlates of dominance (including aggression against intruders, mating, and tube dominance) as well as novelty-seeking and risk-taking responses. Furthermore, male 5αR2 KO mice exhibited reduced D2-like dopamine receptor binding in the shell of the nucleus accumbens - a well-recognized molecular signature of social dominance. Collectively, these results suggest that 5αR2 is involved in the establishment of social dominance and its behavioral manifestations. Further studies are warranted to understand how the metabolic actions of 5αR2 on steroid profile may be implicated in social ranking, impulse control, and the modulation of dopamine receptor expression in the nucleus accumbens.
Collapse
Affiliation(s)
- Laura J Mosher
- Dept of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, United States; Dept of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Sean C Godar
- Dept of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, United States
| | - Marc Morissette
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Quebec, Canada
| | - Kenneth M McFarlin
- Dept of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States.
| | - Simona Scheggi
- Dept of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, United States; Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Carla Gambarana
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Stephen C Fowler
- Dept of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Thérèse Di Paolo
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Quebec, Canada; Faculty of Pharmacy, Laval University, Quebec City, Quebec, Canada
| | - Marco Bortolato
- Dept of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
25
|
Hepatoprotective Effects of Kaempferol-3-O-α-l-Arabinopyranosyl-7-O-α-l-Rhamnopyranoside on d-Galactosamine and Lipopolysaccharide Caused Hepatic Failure in Mice. Molecules 2017; 22:molecules22101755. [PMID: 29057809 PMCID: PMC6151520 DOI: 10.3390/molecules22101755] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/10/2017] [Accepted: 10/13/2017] [Indexed: 12/30/2022] Open
Abstract
Fulminant hepatic failure (FHF), associated with high mortality, is characterized by extensive death of hepatocytes and hepatic dysfunction. There is no effective treatment for FHF. Several studies have indicated that flavonoids can protect the liver from different factor-induced injury. Previously, we found that the extracts of Elaeagnus mollis leaves had favorable protective effects on acute liver injury. However, the role and mechanisms behind that was elusive. This study examined the hepatoprotective mechanisms of kaempferol-3-O-α-l-arabinopyranosyl-7-O-α-l-rhamnopyra-noside (KAR), a major flavonol glycoside of E. mollis, against d-galactosamine (GalN) and lipopolysaccharide (LPS)-induced hepatic failure. KAR reduces the mouse mortality, protects the normal liver structure, inhibits the serum aspartate aminotransferase (AST) and alamine aminotransferase (ALT) activity and decreases the production of malondialdehyde (MDA) and reactive oxygen species (ROS) and inflammatory cytokines, TNF-α, IL-6, and IL-1β. Furthermore, KAR inhibits the apoptosis of hepatocytes and reduces the expression of TLR4 and NF-κB signaling pathway-related proteins induced by GalN/LPS treatment. These findings suggest that the anti-oxidative, anti-inflammatory, and anti-apoptotic effects of KAR on GalN/LPS-induced acute liver injury were performed through down-regulating the activity of the TLR4 and NF-κB signaling pathways.
Collapse
|
26
|
Allopregnanolone mediates the exacerbation of Tourette-like responses by acute stress in mouse models. Sci Rep 2017; 7:3348. [PMID: 28611376 PMCID: PMC5469807 DOI: 10.1038/s41598-017-03649-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/02/2017] [Indexed: 12/26/2022] Open
Abstract
Tourette syndrome (TS) is a neuropsychiatric disorder characterized by multiple tics and sensorimotor abnormalities, the severity of which is typically increased by stress. The neurobiological underpinnings of this exacerbation, however, remain elusive. We recently reported that spatial confinement (SC), a moderate environmental stressor, increases tic-like responses and elicits TS-like sensorimotor gating deficits in the D1CT-7 mouse, one of the best-validated models of TS. Here, we hypothesized that these adverse effects may be mediated by neurosteroids, given their well-documented role in stress-response orchestration. Indeed, SC increased the levels of progesterone, as well as its derivatives 5α-dihydroprogesterone and allopregnanolone, in the prefrontal cortex (PFC) of D1CT-7 mice. Among these steroids, however, only allopregnanolone (5-15 mg/kg, IP) dose-dependently exacerbated TS-like manifestations in D1CT-7, but not wild-type littermates; these effects were countered by the benchmark anti-tic therapy haloperidol (0.3 mg/kg, IP). Furthermore, the phenotypic effects of spatial confinement in D1CT-7 mice were suppressed by finasteride (25-50 mg/kg, IP), an inhibitor of the main rate-limiting enzyme in allopregnanolone synthesis. These findings collectively suggest that stress may exacerbate TS symptoms by promoting allopregnanolone synthesis in the PFC, and corroborate previous clinical results pointing to finasteride as a novel therapeutic avenue to curb symptom fluctuations in TS.
Collapse
|