1
|
Shapouri S, Sharifi A, Folkedal O, Fraser TWK, Vindas MA. Behavioral and neurophysiological effects of buspirone in healthy and depression-like state juvenile salmon. Front Behav Neurosci 2024; 18:1285413. [PMID: 38410095 PMCID: PMC10894974 DOI: 10.3389/fnbeh.2024.1285413] [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: 08/30/2023] [Accepted: 01/11/2024] [Indexed: 02/28/2024] Open
Abstract
A proportion of farmed salmon in seawater show a behaviorally inhibited, growth stunted profile known as a depression-like state (DLS). These DLS fish are characterized by chronically elevated serotonergic signaling and blood plasma cortisol levels and the inability to react further to acute stress, which is suggestive of chronic stress. In this study, we characterize the neuroendocrine profile of growth stunted freshwater parr and confirm that they show a DLS-like neuroendocrine profile with a blunted cortisol response and no serotonergic increase in response to acute stress. Furthermore, we attempted to reverse this DLS-like profile through pharmacological manipulation of the serotonin (5-HT) system with buspirone, an anxiolytic medication that acts as a serotonin receptor agonist (i.e., decreases serotonergic signaling). We found that while buspirone decreases anxiolytic-type behavior in healthy fish, no quantifiable behavioral change was found in DLS-like fish. However, there was a physiological effect of diminished basal serotonergic signaling. This suggests that at the physiological level, buspirone appears to reverse the neuroendocrine DLS profile. With a deeper understanding of what causes DLS profiles and growth stunting in juvenile fish, steps can be taken in terms of husbandry to prevent repeated stressors and the formation of the DLS profile, potentially reducing losses in aquaculture due to chronic stress.
Collapse
Affiliation(s)
- Sheyda Shapouri
- Biochemistry and Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, As, Norway
| | - Aziz Sharifi
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Ole Folkedal
- Animal Welfare, Matre Research Station, Institute of Marine Research, Bergen, Norway
| | - Thomas W. K. Fraser
- Reproduction and Developmental Biology, Matre Research Station, Institute of Marine Research, Bergen, Norway
| | - Marco A. Vindas
- Biochemistry and Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, As, Norway
| |
Collapse
|
2
|
Rodnyy AY, Kondaurova EM, Tsybko AS, Popova NK, Kudlay DA, Naumenko VS. The brain serotonin system in autism. Rev Neurosci 2024; 35:1-20. [PMID: 37415576 DOI: 10.1515/revneuro-2023-0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/10/2023] [Indexed: 07/08/2023]
Abstract
Autism spectrum disorders (ASDs) are among the most common neurodevelopmental diseases. These disorders are characterized by lack of social interaction, by repetitive behavior, and often anxiety and learning disabilities. The brain serotonin (5-HT) system is known to be crucially implicated in a wide range of physiological functions and in the control of different kinds of normal and pathological behavior. A growing number of studies indicate the involvement of the brain 5-HT system in the mechanisms underlying both ASD development and ASD-related behavioral disorders. There are some review papers describing the role of separate key players of the 5-HT system in an ASD and/or autistic-like behavior. In this review, we summarize existing data on the participation of all members of the brain 5-HT system, namely, 5-HT transporter, tryptophan hydroxylase 2, MAOA, and 5-HT receptors, in autism in human and various animal models. Additionally, we describe the most recent studies involving modern techniques for in vivo regulation of gene expression that are aimed at identifying exact roles of 5-HT receptors, MAOA, and 5-HT transporter in the mechanisms underlying autistic-like behavior. Altogether, results of multiple research articles show that the brain 5-HT system intimately partakes in the control of some types of ASD-related behavior, and that specific changes in a function of a certain 5-HT receptor, transporter, and/or enzyme may normalize this aberrant behavior. These data give hope that some of clinically used 5-HT-related drugs have potential for ASD treatment.
Collapse
Affiliation(s)
- Alexander Ya Rodnyy
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Akad. Lavrentyeva Ave. 10, Novosibirsk 630090, Russia
| | - Elena M Kondaurova
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Akad. Lavrentyeva Ave. 10, Novosibirsk 630090, Russia
| | - Anton S Tsybko
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Akad. Lavrentyeva Ave. 10, Novosibirsk 630090, Russia
| | - Nina K Popova
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Akad. Lavrentyeva Ave. 10, Novosibirsk 630090, Russia
| | - Dmitry A Kudlay
- NRC Institute of Immunology FMBA of Russia, Kashirskoe Highway 24, Moscow 115522, Russia
- Sechenov's University, 8-2 Trubetskaya Str., Moscow 119991, Russia
| | - Vladimir S Naumenko
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Akad. Lavrentyeva Ave. 10, Novosibirsk 630090, Russia
| |
Collapse
|
3
|
Calapai F, Mannucci C, McQuain L, Salvo F. Pharmacological Evaluation of Signals of Disproportionality Reporting Related to Adverse Reactions to Antiepileptic Cannabidiol in VigiBase. Pharmaceuticals (Basel) 2023; 16:1420. [PMID: 37895891 PMCID: PMC10610535 DOI: 10.3390/ph16101420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/11/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Cannabidiol is the first cannabis-derived drug approved for the treatment of Lennox-Gastaut syndrome, Dravet syndrome, and Tuberous Sclerosis Complex. In the current study, we performed a descriptive analysis followed by a disproportionality analysis of potential adverse events caused by CBD extracted from the VigiBase® database. Furthermore, the biological plausibility of the association between CBD and the serotonin 5-HT1A receptor as a possible cause of adverse events was analyzed and discussed. Data were extracted from the VigiBase® database using the VigiLyze® signal detection and signal management tool. Adverse events in VigiBase® reports were coded using MedDRA, version 19 of Preferred Terms (PTs). Data were uploaded into SPSS software and analyzed via a disproportionality analysis. Statistically significant disproportionality signals for CBD were found for "weight decreased" (5.19 (95% CI: 4.54-5.70)), "hypophagia" (3.68 (95% CI: 3.22-5.27)), and "insomnia" (1.6 (95% CI: 1.40-1.83)). Positive IC025 values were found for "weight decreased" (2.2), "hypophagia" (1.3), and "insomnia" (0.5), indicating a surplus of reported cases. CBD's interactions with 5-HT1A serotonin receptors may offer a potential biological explanation for the occurrence of insomnia in patients. It is noteworthy that the risk profiles mentioned in the information for prescribing CBD as an antiepileptic agent by regulatory agencies showed disparities specifically related to the adverse event "insomnia".
Collapse
Affiliation(s)
- Fabrizio Calapai
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98125 Messina, Italy;
| | - Carmen Mannucci
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, 98125 Messina, Italy
| | - Liana McQuain
- Université de Bordeaux, European Training Programme in Pharmacovigilance and Pharmacoepidemiology (Eu2P), F-33000 Bordeaux, France;
| | - Francesco Salvo
- CHU de Bordeaux, Service de Pharmacologie Médicale, Centre Régional de Pharmacovigilance de Bordeaux, F-33000 Bordeaux, France;
- Université de Bordeaux, INSERM, Bordeaux Population Health, U1219, AHeaD Team, F-33000 Bordeaux, France
| |
Collapse
|
4
|
On the role of serotonin 5-HT 1A receptor in autistic-like behavior: сross talk of 5-HT and BDNF systems. Behav Brain Res 2023; 438:114168. [PMID: 36280010 DOI: 10.1016/j.bbr.2022.114168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 12/05/2022]
Abstract
Autism spectrum disorders (ASDs) are some of the most common neurodevelopmental disorders; however, the mechanisms underlying ASDs are still poorly understood. Serotonin (5-HT) and brain-derived neurotrophic factor (BDNF) are known as key players in brain and behavioral plasticity and interact with each other. 5-HT1A receptor is a principal regulator of the brain 5-HT system, which modulates normal and pathological behavior. Here we investigated effects of adeno-associated-virus-based 5-HT1A receptor overexpression in the hippocampus of BTBR mice (which are a model of autism) on various types of behavior and on the expression of 5-HT7 receptor, proBDNF, mature BDNF, and BDNF receptors (TrkB and p75NTR). The 5-HT1A receptor overexpression in BTBR mice reduced stereotyped behavior in the marble-burying test and extended the time spent in the center in the open field test. Meanwhile, this overexpression failed to affect social behavior in the three-chambered test, immobility time in the tail suspension test, locomotor activity in the open field test, and associative learning within the "operant wall" paradigm. The 5-HT1A receptor overexpression in the hippocampus raised hippocampal 5-HT7 receptor mRNA and protein levels. Additionally, the 5-HT1A receptor overexpression lowered both mRNA and protein levels of TrkB receptor but failed to affect proBDNF, mature BDNF, and p75NTR receptor expression in the hippocampus of BTBR mice. Thus, obtained results suggest the involvement of the 5-HT and BDNF systems' interaction mediated by 5-HT1A and TrkB receptors in the mechanisms underlying autistic-like behavior in BTBR mice.
Collapse
|
5
|
Wang C, Yang S, Deng J, Shi L, Chang J, Meng J, Liu W, Zeng J, Xing K, Wen J, Liang B, Xing D. The research progress on the anxiolytic effect of plant-derived flavonoids by regulating neurotransmitters. Drug Dev Res 2023; 84:458-469. [PMID: 36744648 DOI: 10.1002/ddr.22038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/30/2022] [Accepted: 01/13/2023] [Indexed: 02/07/2023]
Abstract
Phytopharmaceuticals have attracted a lot of attention due to their multicomponent and multiple targets. The natural phenolic chemicals known as flavonoids are found in a wide variety of plants, fruits, vegetables, and herbs. Recently, they have been found to have modulatory effects on anxiety disorders, with current research focusing on the modulation of neurotransmitters. There has not yet been a review of the various natural flavonoid monomer compounds and total plant flavonoids that have been found to have anxiolytic effects. The study on the anti-anxiety effects of plant-derived flavonoids on neurotransmitters was reviewed in this paper. We, therefore, anticipate that further study on the conformational interaction underlying flavonoids' anti-anxiety effects will offer a theoretical framework for the creation of pertinent treatments.
Collapse
Affiliation(s)
- Chao Wang
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, Shandong, China
| | - Shanbo Yang
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, Shandong, China.,School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Junwen Deng
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, Shandong, China.,School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Lingyu Shi
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, Shandong, China.,School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Jing Chang
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, Shandong, China.,School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Jingsen Meng
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, Shandong, China.,School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Wenjing Liu
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, Shandong, China.,School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Jun Zeng
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, Shandong, China.,School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Kunyue Xing
- Alliance Manchester Business School, The University of Manchester, Manchester, UK
| | - Jialian Wen
- School of Social Science, The University of Manchester, Manchester, UK
| | - Bing Liang
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, Shandong, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, Shandong, China.,School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
6
|
Powell WH, Annett LE, Depoortere R, Newman-Tancredi A, Iravani MM. The selective 5-HT 1A receptor agonist NLX-112 displays anxiolytic-like activity in mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:149-157. [PMID: 34821958 DOI: 10.1007/s00210-021-02183-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/10/2021] [Indexed: 10/19/2022]
Abstract
Anxiety is amongst the commonest neuropsychiatric disorders, and there is a large body of evidence to suggest that abnormalities in serotonergic function are involved in its pathogenesis. Several studies have implicated 5-HT1A receptor activation in mitigating anxiety disorders, so this study investigated the acute effects of a highly selective, potent and efficacious 5-HT1A receptor full agonist, NLX-112 (a.k.a. befiradol, F13640), in middle-aged C57bl/6 J male mice. Video tracking was used to measure several parameters including time spent in the open and closed arms of an elevated plus maze (EPM), distance travelled and thigmotaxis in an open field test (OFT). At 0.1 to 1.0 mg/kg s.c., NLX-112 markedly decreased thigmotaxis and increased exploratory behaviour in the OFT and EPM assays. Hence, at 0.3 mg/kg, NLX-112 augmented locomotor activity in the centre of an open field arena by 164% and increased the time spent in the open arms of the EPM by 119% of control. These results indicate that anxiety-like behaviours in mice are significantly diminished with low doses of NLX-112. NLX-112 may therefore possess anxiolytic properties which complement its known activity in models of movement disorders.
Collapse
Affiliation(s)
- William H Powell
- Department of Clinical Pharmaceutical and Biological Science, University of Hertfordshire, Hatfield, UK
| | - Lucy E Annett
- Department of Psychology, School of Life and Medical Sciences, College Lane, University of Hertfordshire, Hatfield, AL10 9AB, UK
| | | | | | - Mahmoud M Iravani
- Department of Clinical Pharmaceutical and Biological Science, University of Hertfordshire, Hatfield, UK.
| |
Collapse
|
7
|
Voronova IP. 5-HT Receptors and Temperature Homeostasis. Biomolecules 2021; 11:1914. [PMID: 34944557 PMCID: PMC8699715 DOI: 10.3390/biom11121914] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/11/2021] [Accepted: 12/16/2021] [Indexed: 12/28/2022] Open
Abstract
The present review summarizes the data concerning the influence of serotonin (5-HT) receptors on body temperature in warm-blooded animals and on processes associated with its maintenance. This review includes the most important part of investigations from the first studies to the latest ones. The established results on the pharmacological activation of 5-HT1A, 5-HT3, 5-HT7 and 5-HT2 receptor types are discussed. Such activation of the first 3 type of receptors causes a decrease in body temperature, whereas the 5-HT2 activation causes its increase. Physiological mechanisms leading to changes in body temperature as a result of 5-HT receptors' activation are discussed. In case of 5-HT1A receptor, they include an inhibition of shivering and non-shivering thermogenesis, as well simultaneous increase of peripheral blood flow, i.e., the processes of heat production and heat loss. The physiological processes mediated by 5-HT2 receptor are opposite to those of the 5-HT1A receptor. Mechanisms of 5-HT3 and 5-HT7 receptor participation in these processes are yet to be studied in more detail. Some facts indicating that in natural conditions, without pharmacological impact, these 5-HT receptors are important links in the system of temperature homeostasis, are also discussed.
Collapse
Affiliation(s)
- Irina P. Voronova
- Department of Thermophysiology, Scientific Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia
| |
Collapse
|
8
|
Murkar A, De Koninck J, Merali Z. Cannabinoids: Revealing their complexity and role in central networks of fear and anxiety. Neurosci Biobehav Rev 2021; 131:30-46. [PMID: 34487746 DOI: 10.1016/j.neubiorev.2021.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 12/11/2022]
Abstract
The first aim of the present review is to provide an in-depth description of the cannabinoids and their known effects at various neuronal receptors. It reveals that cannabinoids are highly diverse, and recent work has highlighted that their effects on the central nervous system (CNS) are surprisingly more complex than previously recognized. Cannabinoid-sensitive receptors are widely distributed throughout the CNS where they act as primary modulators of neurotransmission. Secondly, we examine the role of cannabinoid receptors at key brain sites in the control of fear and anxiety. While our understanding of how cannabinoids specifically modulate these networks is mired by their complex interactions and diversity, a plausible framework(s) for their effects is proposed. Finally, we highlight some important knowledge gaps in our understanding of the mechanism(s) responsible for their effects on fear and anxiety in animal models and their use as therapeutic targets in humans. This is particularly important for our understanding of the phytocannabinoids used as novel clinical interventions.
Collapse
Affiliation(s)
- Anthony Murkar
- University of Ottawa Institute of Mental Health Research (IMHR), Ottawa, ON, Canada; School of Psychology, University of Ottawa, Ottawa, ON, Canada.
| | - Joseph De Koninck
- University of Ottawa Institute of Mental Health Research (IMHR), Ottawa, ON, Canada; School of Psychology, University of Ottawa, Ottawa, ON, Canada
| | - Zul Merali
- School of Psychology, University of Ottawa, Ottawa, ON, Canada; Brain and Mind Institute, Aga Khan University, Nairobi, Kenya; Carleton University, Neuroscience Department, Ottawa, ON, Canada
| |
Collapse
|
9
|
Ozawa A, Arakawa H. Chemogenetics drives paradigm change in the investigation of behavioral circuits and neural mechanisms underlying drug action. Behav Brain Res 2021; 406:113234. [PMID: 33741409 PMCID: PMC8110310 DOI: 10.1016/j.bbr.2021.113234] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
Recent developments in chemogenetic approaches to the investigation of brain function have ushered in a paradigm change in the strategy for drug and behavior research and clinical drug-based medications. As the nature of the drug action is based on humoral regulation, it is a challenge to identify the neuronal mechanisms responsible for the expression of certain targeted behavior induced by drug application. The development of chemogenetic approaches has allowed researchers to control neural activities in targeted neurons through a toolbox, including engineered G protein-coupled receptors or ligand-gated ion channels together with exogenously inert synthetic ligands. This review provides a brief overview of the chemogenetics toolbox with an emphasis on the DREADDs (Designer Receptors Exclusively Activated by Designer Drugs) technique used in rodent models, which is applicable to the investigation of how specific neural circuits regulate behavioral processes. The use of chemogenetics has had a significant impact on basic neuroscience for a better understanding of the relationships between brain activity and the expression of behaviors with cell- and circuit-specific orders. Furthermore, chemogenetics is potentially a useful tool to deconstruct the neuropathological mechanisms of mental diseases and its regulation by drug, and provide us with transformative therapeutics with medication. We also review recent findings in the use of chemogenetic techniques to uncover functional circuit connections of serotonergic neurons in rodent models.
Collapse
Affiliation(s)
- Akihiko Ozawa
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL, USA
| | - Hiroyuki Arakawa
- Department of Psychology, Tokiwa University, Mito, Ibaraki, Japan; Department of Systems Physiology, University of Ryukyus, Faculty of Medicine, Nakagami District, Okinawa, Japan.
| |
Collapse
|
10
|
Echeverry C, Prunell G, Narbondo C, de Medina VS, Nadal X, Reyes-Parada M, Scorza C. A Comparative In Vitro Study of the Neuroprotective Effect Induced by Cannabidiol, Cannabigerol, and Their Respective Acid Forms: Relevance of the 5-HT 1A Receptors. Neurotox Res 2021; 39:335-348. [PMID: 32886342 DOI: 10.1007/s12640-020-00277-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/11/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022]
Abstract
Previous preclinical studies have demonstrated that cannabidiol (CBD) and cannabigerol (CBG), two non-psychotomimetic phytocannabinoids from Cannabis sativa, induce neuroprotective effects on toxic and neurodegenerative processes. However, a comparative study of both compounds has not been reported so far, and the targets involved in this effect remain unknown. The ability of CBD and CBG to attenuate the neurotoxicity induced by two insults involving oxidative stress (hydrogen peroxide, H2O2) and mitochondrial dysfunction (rotenone) was evaluated in neural cell cultures. The involvement of CB-1 and CB-2 or 5-HT1A receptors was investigated. The neuroprotective effect of their respective acids forms, cannabidiolic acid (CBDA) and cannabigerolic acid (CBGA), was also analyzed. MTT and immunocytochemistry assays were used to evaluate cell viability. No significant variation on cell viability was per se induced by the lower concentrations tested of CBD and CBG or CBDA and CBGA; however, high concentrations of CBD, CBDA, or CBGA were toxic since a 40-50% reduction of cell viability was observed. CBD and CBG showed neuroprotective effects against H2O2 or rotenone; however, both compounds were more effective in attenuating the rotenone-induced neurotoxicity. A high concentration of CBDA reduced the rotenone-induced neurotoxicity. WAY100635 (5-HT1A receptor antagonist) but not AM251 and AM630 (CB1 or CB2 receptor antagonists, respectively) significantly diminished the neuroprotective effect induced by CBG only against rotenone. Our results contribute to the understanding of the neuroprotective effect of CBD and CBG, showing differences with their acid forms, and also highlight the role of 5-HT1A receptors in the mechanisms of action of CBG.
Collapse
Affiliation(s)
- Carolina Echeverry
- Department of Neurochemistry, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Avenida Italia 3318, CP 11600, Montevideo, Uruguay.
| | - Giselle Prunell
- Department of Neurochemistry, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Avenida Italia 3318, CP 11600, Montevideo, Uruguay
| | - Camila Narbondo
- Department of Neurochemistry, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Avenida Italia 3318, CP 11600, Montevideo, Uruguay
| | | | - Xavier Nadal
- EthnoPhytoTech Research & Consulting S.L.U., Sant Cugat del Valles, Spain
| | - Miguel Reyes-Parada
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago, Chile
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Cecilia Scorza
- Department of Experimental Neuropharmacology, IIBCE, Montevideo, Uruguay
| |
Collapse
|
11
|
Luo OD, Kwiecien-Delaney B, Martin P, Foster JA, Sidor MM. The effect of early life immune challenge on adult forced swim test performance and hippocampal neurogenesis. J Neuroimmunol 2021; 354:577530. [PMID: 33744708 DOI: 10.1016/j.jneuroim.2021.577530] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 10/22/2022]
Abstract
Many psychiatric diseases can be considered neurodevelopmental in nature and accumulating evidence links immune system dysfunction to disease etiology. Yet, it is currently unknown how the immune system alters brain function through development to increase susceptibility to psychiatric illness. Neonatal immune challenge in rodents is a neurodevelopmental model that has been associated with long-term molecular and behavioural changes in stress-reactivity. As enhanced stress-reactivity is associated with the emergence of depressive-like behaviours concurrent with hippocampal pathology, we measured depressive-like behaviour in the forced swim test and hippocampal neurogenesis in adult mice neonatally exposed to lipopolysaccharide LPS; 0.05 mg/kg, i.p. on postnatal days 3 and 5. As there are important functional differences along the ventral-dorsal hippocampus axis, ventral and dorsal hippocampal neurogenesis were measured separately. Our findings reveal a sexually-dimorphic response to early-life LPS challenge. Male LPS-mice spent less time immobile in the forced swim test, suggesting altered reactivity to swim stress. This was accompanied by an increase in doublecortin-positive cells in the dorsal hippocampus of female mice. These findings demonstrate that exposure to an immune challenge during critical developmental time periods leads to long-term sexually-dimorphic alterations in stress-reactivity that are accompanied by changes to adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Owen D Luo
- Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, Ontario, Canada
| | | | - Patrick Martin
- Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, Ontario, Canada
| | - Jane A Foster
- Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, Ontario, Canada; Department of Psychiatry, St. Michael's Hospital, Toronto, Ontario, Canada.
| | - Michelle M Sidor
- Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
12
|
Mechtler LL, Gengo FM, Bargnes VH. Cannabis and Migraine: It's Complicated. Curr Pain Headache Rep 2021; 25:16. [PMID: 33630181 DOI: 10.1007/s11916-020-00931-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2020] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The use of cannabis for the treatment of migraine has become an area of interest with the legalization of medical cannabis in the USA. Understanding the mechanisms of cannabinoids, available studies, and best clinical recommendations is crucial for headache providers to best serve patients. RECENT FINDINGS Patients utilizing medical cannabis for migraine have reported improvement in migraine profile and common comorbidities. Reduction in prescription medication is also common, especially opioids. Side effects exist, with the majority being mild. Not enough data is available for specific dose recommendations, but THC and CBD appear to mediate these observed effects. The purpose of this article is twofold: review the limited research surrounding cannabis for migraine disease and reflect on clinical management experiences to provide recommendations that best capture the potential use of cannabis for migraine.
Collapse
Affiliation(s)
- Laszlo L Mechtler
- Dent Neurologic Institute, 3980 Sheridan Drive, Suite 600, Amherst, NY, 14226, USA.
| | - Fran M Gengo
- Dent Neurologic Institute, 3980 Sheridan Drive, Suite 300, Amherst, NY, 14226, USA
| | - Vincent H Bargnes
- Dent Neurologic Institute, 3980 Sheridan Drive, Suite 600, Amherst, NY, 14226, USA
| |
Collapse
|
13
|
Cannabidiol as a Potential Treatment for Anxiety and Mood Disorders: Molecular Targets and Epigenetic Insights from Preclinical Research. Int J Mol Sci 2021; 22:ijms22041863. [PMID: 33668469 PMCID: PMC7917759 DOI: 10.3390/ijms22041863] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 02/08/2023] Open
Abstract
Cannabidiol (CBD) is the most abundant non-psychoactive component of cannabis; it displays a very low affinity for cannabinoid receptors, facilitates endocannabinoid signaling by inhibiting the hydrolysis of anandamide, and stimulates both transient receptor potential vanilloid 1 and 2 and serotonin type 1A receptors. Since CBD interacts with a wide variety of molecular targets in the brain, its therapeutic potential has been investigated in a number of neuropsychiatric diseases, including anxiety and mood disorders. Specifically, CBD has received growing attention due to its anxiolytic and antidepressant properties. As a consequence, and given its safety profile, CBD is considered a promising new agent in the treatment of anxiety and mood disorders. However, the exact molecular mechanism of action of CBD still remains unknown. In the present preclinical review, we provide a summary of animal-based studies that support the use of CBD as an anxiolytic- and antidepressant-like compound. Next, we describe neuropharmacological evidence that links the molecular pharmacology of CBD to its behavioral effects. Finally, by taking into consideration the effects of CBD on DNA methylation, histone modifications, and microRNAs, we elaborate on the putative role of epigenetic mechanisms in mediating CBD’s therapeutic outcomes.
Collapse
|
14
|
Arakawa H. Dynamic regulation of oxytocin neuronal circuits in the sequential processes of prosocial behavior in rodent models. CURRENT RESEARCH IN NEUROBIOLOGY 2021; 2:100011. [PMID: 36246512 PMCID: PMC9559098 DOI: 10.1016/j.crneur.2021.100011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/08/2021] [Accepted: 03/31/2021] [Indexed: 12/16/2022] Open
|
15
|
Jia T, Cao L, Ye X, Wei Q, Xie W, Cai C, Dong Y, Xia C, Tian Y, Wang K. Difference in binocular rivalry rate between major depressive disorder and generalized anxiety disorder. Behav Brain Res 2020; 391:112704. [PMID: 32461128 DOI: 10.1016/j.bbr.2020.112704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 04/28/2020] [Accepted: 05/12/2020] [Indexed: 10/24/2022]
Abstract
Binocular rivalry (BR) occurs when monocular images are presented simultaneously to corresponding regions of the two eyes. Rather than forming a stable composite, perception alternates spontaneously between each monocular view. Over recent decades, considerable interest in examining BR in psychiatric populations has demonstrated clear differences in the dynamics of BR when compared to healthy populations. However, the available data with respect to rivalry rates in depression and anxiety are limited, and previous studies have shown inconsistent results. Also, depression and anxiety are highly comorbid and can be difficult to distinguish. Therefore, we aimed to investigate whether patients with depression and anxiety have abnormal rivalry rates and whether rivalry rates differ between these conditions. Thirty-five patients with major depressive disorder (MDD), 30 patients with generalized anxiety disorder (GAD), and 30 control participants matched on sex, age and education were recruited. Our results showed MDD participants had slower BR rates than controls and GAD, and controls were significantly slower than GAD. Our findings raise prospects that BR could be a promising behavioral tool to differentiate depressive and anxiety disorders.
Collapse
Affiliation(s)
- Ting Jia
- Department of Neurology, Nanjing Pukou Central Hospital, Nanjing, Jiangsu Province, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Lihua Cao
- Department of Neurology, Nanjing Pukou Central Hospital, Nanjing, Jiangsu Province, China
| | - Xing Ye
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Qiang Wei
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Wen Xie
- Department of Psychiatry, Anhui Mental Health Center, Hefei, Anhui Province, China
| | - Chunlan Cai
- Department of Psychiatry, Anhui Mental Health Center, Hefei, Anhui Province, China
| | - Yi Dong
- Department of Psychiatry, Anhui Mental Health Center, Hefei, Anhui Province, China
| | - Chengcai Xia
- Department of Neurology, Nanjing Pukou Central Hospital, Nanjing, Jiangsu Province, China
| | - Yanghua Tian
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China.
| | - Kai Wang
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China.
| |
Collapse
|
16
|
Dey D, Parihar VK, Szabo GG, Klein PM, Tran J, Moayyad J, Ahmed F, Nguyen QA, Murry A, Merriott D, Nguyen B, Goldman J, Angulo MC, Piomelli D, Soltesz I, Baulch JE, Limoli CL. Neurological Impairments in Mice Subjected to Irradiation and Chemotherapy. Radiat Res 2020; 193:407-424. [PMID: 32134362 DOI: 10.1667/rr15540.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Radiotherapy, surgery and the chemotherapeutic agent temozolomide (TMZ) are frontline treatments for glioblastoma multiforme (GBM). However beneficial, GBM treatments nevertheless cause anxiety or depression in nearly 50% of patients. To further understand the basis of these neurological complications, we investigated the effects of combined radiotherapy and TMZ chemotherapy (combined treatment) on neurological impairments using a mouse model. Five weeks after combined treatment, mice displayed anxiety-like behaviors, and at 15 weeks both anxiety- and depression-like behaviors were observed. Relevant to the known roles of the serotonin axis in mood disorders, we found that 5HT1A serotonin receptor levels were decreased by ∼50% in the hippocampus at both early and late time points, and a 37% decrease in serotonin levels was observed at 15 weeks postirradiation. Furthermore, chronic treatment with the selective serotonin reuptake inhibitor fluoxetine was sufficient for reversing combined treatment-induced depression-like behaviors. Combined treatment also elicited a transient early increase in activated microglia in the hippocampus, suggesting therapy-induced neuroinflammation that subsided by 15 weeks. Together, the results of this study suggest that interventions targeting the serotonin axis may help ameliorate certain neurological side effects associated with the clinical management of GBM to improve the overall quality of life for cancer patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Faizy Ahmed
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
| | | | | | | | | | | | | | - Daniele Piomelli
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
| | - Ivan Soltesz
- Departments of Neurology and Neurological Sciences, Stanford University, Palo Alto, California 94305
| | | | | |
Collapse
|
17
|
Staes N, Sherwood CC, Freeman H, Brosnan SF, Schapiro SJ, Hopkins WD, Bradley BJ. Serotonin Receptor 1A Variation Is Associated with Anxiety and Agonistic Behavior in Chimpanzees. Mol Biol Evol 2020; 36:1418-1429. [PMID: 31045220 DOI: 10.1093/molbev/msz061] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Serotonin is a neurotransmitter that plays an important role in regulating behavior and personality in humans and other mammals. Polymorphisms in genes coding for the serotonin receptor subtype 1A (HTR1A), the serotonin transporter (SLC6A4), and the serotonin degrading enzyme monoamine oxidase A (MAOA) are associated with anxiety, impulsivity, and neurotic personality in humans. In primates, previous research has largely focused on SLC6A4 and MAOA, with few studies investigating the role of HTR1A polymorphic variation on behavior. Here, we examined variation in the coding region of HTR1A across apes, and genotyped polymorphic coding variation in a sample of 214 chimpanzees with matched measures of personality and behavior. We found evidence for positive selection at three amino acid substitution sites, one in chimpanzees-bonobos (Thr26Ser), one in humans (Phe33Val), and one in orangutans (Ala274Gly). Investigation of the HTR1A coding region in chimpanzees revealed a polymorphic site, where a C/A single nucleotide polymorphism changes a proline to a glutamine in the amino acid sequence (Pro248Gln). The substitution is located in the third intracellular loop of the receptor, a region important for serotonin signal transduction. The derived variant is the major allele in this population (frequency 0.67), and is associated with a reduction in anxiety, decreased rates of male agonistic behavior, and an increase in socio-positive behavior. These results are the first evidence that the HTR1A gene may be involved in regulating social behavior in chimpanzees and encourage further systematic investigation of polymorphic variation in other primate populations with corresponding data on behavior.
Collapse
Affiliation(s)
- Nicky Staes
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC.,Department of Biology, Behavioral Ecology and Ecophysiology Group, University of Antwerp, Antwerp, Belgium.,Centre for Research and Conservation, Royal Zoological Society of Antwerp, Antwerp, Belgium
| | - Chet C Sherwood
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC
| | - Hani Freeman
- Michale E. Keeling Center for Comparative Medicine and Research, The University of Texas MD Anderson Cancer CenterBastrop, TX
| | - Sarah F Brosnan
- Michale E. Keeling Center for Comparative Medicine and Research, The University of Texas MD Anderson Cancer CenterBastrop, TX.,Department of Psychology, Georgia State University, Atlanta, GA.,Neuroscience Institute and Language Research Center, Georgia State University, Atlanta, GA
| | - Steven J Schapiro
- Michale E. Keeling Center for Comparative Medicine and Research, The University of Texas MD Anderson Cancer CenterBastrop, TX
| | - William D Hopkins
- Neuroscience Institute and Language Research Center, Georgia State University, Atlanta, GA.,Ape Cognition and Conservation Initiative, Des Moines, IA
| | - Brenda J Bradley
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC
| |
Collapse
|
18
|
Bourin M. Experimental Anxiety Model for Anxiety Disorders: Relevance to Drug Discovery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1191:169-184. [DOI: 10.1007/978-981-32-9705-0_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
19
|
Zhu LJ, Xu C, Ren J, Chang L, Zhu XH, Sun N, Meng GL, Liu MY, Zhang J, Li YY, Tang YL, Zhou QG. Dentate nNOS accounts for stress-induced 5-HT 1A receptor deficiency: Implication in anxiety behaviors. CNS Neurosci Ther 2019; 26:453-464. [PMID: 31863649 PMCID: PMC7080430 DOI: 10.1111/cns.13269] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 10/28/2019] [Accepted: 10/31/2019] [Indexed: 01/09/2023] Open
Abstract
Background Anxiety is a common disorder with high social burden worldwide. Dysfunction of serotonin‐1A receptor (5‐HT1A receptor) in the dentate gyrus (DG) of the hippocampus has been predominantly implicated in the anxiety behavior. However, the molecular mechanism underlying the deficiency of postsynaptic 5‐HT1A receptor in regulating anxiety behavior remains unclear. Methods Using pharmacological and genetic methods, we investigated the role of detate nNOS in 5‐HT1A receptor decline and anxiety behavior induced by chronic mild stress (CMS) in mice. Results Here we showed that local elevation of glucocorticoids in the DG accounted for chronic stress‐induced anxiety behavior. Neuronal nitric oxide synthase (nNOS) mediated chronic stress‐induced downregulation of 5‐HT1A receptor in the DG through peroxynitrite anion (ONOO•) pathway but not cyclic guanosine monophosphate (cGMP) pathway. By using pharmacological tool drugs and nNOS knockout mice, we found that nNOS in the DG played a key role in chronic stress‐induced anxiety behavior. Conclusions These findings uncovered an important role of nNOS‐5‐HT1A receptor pathway in the DG of the hippocampus in chronic stress‐induced anxiety. Accordingly, we developed a “dentate nNOS‐5‐HT1A receptor closed‐loop” theory (stress‐glucocorticoids‐nNOS‐Nitric oxide‐ONOO•‐5‐HT1A receptor ‐nNOS) of stress‐related anxiety.
Collapse
Affiliation(s)
- Li-Juan Zhu
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China.,Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Institute of Neuroscience, Soochow University, Suzhou, China
| | - Chu Xu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Jie Ren
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| | - Lei Chang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xian-Hui Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmcay, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Nan Sun
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Guo-Liang Meng
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Meng-Ying Liu
- Department of Pharmacy, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jing Zhang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yuan-Yuan Li
- Department of Clinical Pharmcay, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Yu-Lin Tang
- Department of Clinical Pharmcay, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Qi-Gang Zhou
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Department of Clinical Pharmcay, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
20
|
Budylin T, Guariglia SR, Duran LI, Behring BM, Shaikh Z, Neuwirth LS, Banerjee P. Ultrasonic vocalization sex differences in 5-HT-R deficient mouse pups: Predictive phenotypes associated with later-life anxiety-like behaviors. Behav Brain Res 2019; 373:112062. [DOI: 10.1016/j.bbr.2019.112062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 06/24/2019] [Accepted: 06/25/2019] [Indexed: 01/26/2023]
|
21
|
Gene knockout animal models of depression, anxiety and obsessive compulsive disorders. Psychiatr Genet 2019; 29:191-199. [DOI: 10.1097/ypg.0000000000000238] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
22
|
Kalinichenko LS, Kornhuber J, Müller CP. Individual differences in inflammatory and oxidative mechanisms of stress-related mood disorders. Front Neuroendocrinol 2019; 55:100783. [PMID: 31415777 DOI: 10.1016/j.yfrne.2019.100783] [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: 01/29/2019] [Revised: 08/05/2019] [Accepted: 08/09/2019] [Indexed: 12/16/2022]
Abstract
Emotional stress leads to the development of peripheral disorders and is recognized as a modifiable risk factor for psychiatric disorders, particularly depression and anxiety. However, not all individuals develop the negative consequences of emotional stress due to different stress coping strategies and resilience to stressful stimuli. In this review, we discuss individual differences in coping styles and the potential mechanisms that contribute to individual vulnerability to stress, such as parameters of the immune system and oxidative state. Initial differences in inflammatory and oxidative processes determine resistance to stress and stress-related disorders via the alteration of neurotransmitter content in the brain and biological fluids. Differences in coping styles may serve as possible predictors of resistance to stress and stress-related disorders, even before stressful conditions. The investigation of natural variabilities in stress resilience may allow the development of new methods for preventive medicine and the personalized treatment of stress-related conditions.
Collapse
Affiliation(s)
- L S Kalinichenko
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany.
| | - J Kornhuber
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - C P Müller
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany
| |
Collapse
|
23
|
Xin F, Fischer E, Krapp C, Krizman EN, Lan Y, Mesaros C, Snyder NW, Bansal A, Robinson MB, Simmons RA, Bartolomei MS. Mice exposed to bisphenol A exhibit depressive-like behavior with neurotransmitter and neuroactive steroid dysfunction. Horm Behav 2018; 102:93-104. [PMID: 29763587 PMCID: PMC6261494 DOI: 10.1016/j.yhbeh.2018.05.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 05/07/2018] [Accepted: 05/11/2018] [Indexed: 11/28/2022]
Abstract
Fetal exposure to endocrine disrupting chemicals (EDCs) has been associated with adverse neurobehavioral outcomes across the lifespan and can persist across multiple generations of offspring. However, the underlying mechanisms driving these changes are not well understood. We investigated the molecular perturbations associated with EDC-induced behavioral changes in first (F1) and second (F2) filial generations, using the model EDC bisphenol A (BPA). C57BL/6J dams were exposed to BPA from preconception until lactation through the diet at doses (10 μg/kg bw/d-lower dose or 10 mg/kg bw/d-upper dose) representative of human exposure levels. As adults, F1 male offspring exhibited increased depressive-like behavior, measured by the forced swim test, while females were unaffected. These behavioral changes were limited to the F1 generation and were not associated with altered maternal care. Transcriptome analysis by RNA-sequencing in F1 control and upper dose BPA-exposed adult male hippocampus revealed neurotransmitter systems as major pathways disrupted by developmental BPA exposure. High performance liquid chromatography demonstrated a male-specific reduction in hippocampal serotonin. Administration of the selective serotonin reuptake inhibitor fluoxetine (20 mg/kg bw) rescued the depressive-like phenotype in males exposed to lower, but not upper, dose BPA, suggesting distinct mechanisms of action for each exposure dose. Finally, high resolution mass spectrometry revealed reduced circulating levels of the neuroactive steroid dehydroepiandrosterone in BPA-exposed males, suggesting another potential mechanism underlying the depressive-like phenotype. Thus, behavioral changes associated with early life BPA exposure may be mediated by sex-specific disruptions in the serotonergic system and/or sex steroid biogenesis in male offspring.
Collapse
Affiliation(s)
- Frances Xin
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erin Fischer
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher Krapp
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elizabeth N Krizman
- Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yemin Lan
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Clementina Mesaros
- Center for Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Cancer Pharmacology, Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nathaniel W Snyder
- A.J. Drexel Autism Institute, Drexel University, Philadelphia, PA 19104, USA
| | - Amita Bansal
- Center for Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Research on Reproduction and Women's Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael B Robinson
- Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rebecca A Simmons
- Center for Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Research on Reproduction and Women's Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marisa S Bartolomei
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
24
|
Kulikov AV, Gainetdinov RR, Ponimaskin E, Kalueff AV, Naumenko VS, Popova NK. Interplay between the key proteins of serotonin system in SSRI antidepressants efficacy. Expert Opin Ther Targets 2018. [DOI: 10.1080/14728222.2018.1452912] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Alexander V. Kulikov
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Allan V. Kalueff
- School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
- Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia
- Ural Federal University, Ekaterinburg 620002, Russia
- Research Institute of Physiology and Basic Medicine, Novosibirsk 630117, Russia
- Russian Research Center for Radiology and Surgical Technologies, Pesochny 197758, Russia
| | - Vladimir S. Naumenko
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Nina K. Popova
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
25
|
Jiménez-Ferrer E, Santillán-Urquiza MA, Alegría-Herrera E, Zamilpa A, Noguerón-Merino C, Tortoriello J, Navarro-García V, Avilés-Flores M, Fuentes-Mata M, Herrera-Ruiz M. Anxiolytic effect of fatty acids and terpenes fraction from Aloysia triphylla: Serotoninergic, GABAergic and glutamatergic implications. Biomed Pharmacother 2017; 96:320-327. [PMID: 29017144 DOI: 10.1016/j.biopha.2017.10.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/29/2017] [Accepted: 10/02/2017] [Indexed: 12/16/2022] Open
Abstract
Aloysia triphylla (Verbenaceae) is an aromatic medicinal plant, and it is used for the treatment of "nervous" problems as, "sadness" and "nervousness". While, there are no reports about its pharmacological activity in animal models. The objective of this work was to evaluate the anxiolytic effect of the extracts and fractions of this species and to measure the interaction of the most active fraction with serotonergic, glutamatergic and GABAergic drugs. An elevated plus maze test was carried ought where the methanol (AtM), dicloromethane (AtD) and hexanic (AtH) extracts presented anxiolytic activity in mice when exposed to the test. Also, different fractions obtained from the AtD were evaluated (AtF1, AtF2 and AtF3, 15mg/kg), and showed that fraction AtF1 possessed the anxiolytic activity, in the same model. Then, AtF1 was co-administered with different drugs, which act on GABAergic (bicuculline, picrotoxin, pentylenetetrazol, baclofen and phaclofen), or serotononinergic (DOI, 8-OH-DPAT, WAY 100635 and ketanserine) or glutamatergic (NMDA, MPEP and MK-801) systems. The anxiolytic activity of AtF1 was modified by GABAergic and serotoninergic drugs. Chemical analysis of this fraction by using GC-MS, showed that it contains hexadecanoic acid, hexadecanoic acid methyl ester, octadecanoic acid methyl ester, eicosanoic acid methyl ester, vitamin E, α-amiryn, campesterol, sitosterol, stigmastan-2,22, dien-3-ol (4) and stigmasta 5, 24 (28) dien-3-ol.
Collapse
Affiliation(s)
- Enrique Jiménez-Ferrer
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina 1, 62790 Xochitepec, Morelos, Mexico
| | - Mayra Alejandra Santillán-Urquiza
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina 1, 62790 Xochitepec, Morelos, Mexico
| | - Elian Alegría-Herrera
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina 1, 62790 Xochitepec, Morelos, Mexico
| | - Alejandro Zamilpa
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina 1, 62790 Xochitepec, Morelos, Mexico
| | - Carmen Noguerón-Merino
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina 1, 62790 Xochitepec, Morelos, Mexico
| | - Jaime Tortoriello
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina 1, 62790 Xochitepec, Morelos, Mexico; INAH-Morelos, Matamoros 14 Acapantzingo, Cuernavaca, Morelos, CP 62440, Mexico
| | - Victor Navarro-García
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina 1, 62790 Xochitepec, Morelos, Mexico
| | | | | | - Maribel Herrera-Ruiz
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina 1, 62790 Xochitepec, Morelos, Mexico.
| |
Collapse
|
26
|
Abstract
Previous attempts to identify a unified theory of brain serotonin function have largely failed to achieve consensus. In this present synthesis, we integrate previous perspectives with new and older data to create a novel bipartite model centred on the view that serotonin neurotransmission enhances two distinct adaptive responses to adversity, mediated in large part by its two most prevalent and researched brain receptors: the 5-HT1A and 5-HT2A receptors. We propose that passive coping (i.e. tolerating a source of stress) is mediated by postsynaptic 5-HT1AR signalling and characterised by stress moderation. Conversely, we argue that active coping (i.e. actively addressing a source of stress) is mediated by 5-HT2AR signalling and characterised by enhanced plasticity (defined as capacity for change). We propose that 5-HT1AR-mediated stress moderation may be the brain's default response to adversity but that an improved ability to change one's situation and/or relationship to it via 5-HT2AR-mediated plasticity may also be important - and increasingly so as the level of adversity reaches a critical point. We propose that the 5-HT1AR pathway is enhanced by conventional 5-HT reuptake blocking antidepressants such as the selective serotonin reuptake inhibitors (SSRIs), whereas the 5-HT2AR pathway is enhanced by 5-HT2AR-agonist psychedelics. This bipartite model purports to explain how different drugs (SSRIs and psychedelics) that modulate the serotonergic system in different ways, can achieve complementary adaptive and potentially therapeutic outcomes.
Collapse
Affiliation(s)
- RL Carhart-Harris
- Psychedelic Research Group, Neuropsychopharmacology Unit, Centre for Psychiatry, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - DJ Nutt
- Psychedelic Research Group, Neuropsychopharmacology Unit, Centre for Psychiatry, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
27
|
Verdurand M, Zimmer L. Hippocampal 5-HT1A receptor expression changes in prodromal stages of Alzheimer's disease: Beneficial or deleterious? Neuropharmacology 2017. [DOI: 10.1016/j.neuropharm.2017.06.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
28
|
Wirth A, Holst K, Ponimaskin E. How serotonin receptors regulate morphogenic signalling in neurons. Prog Neurobiol 2017; 151:35-56. [DOI: 10.1016/j.pneurobio.2016.03.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 03/09/2016] [Accepted: 03/19/2016] [Indexed: 11/25/2022]
|
29
|
Zhang R, Bi Y, Niu W, Huang X, Chen S, Li X, Wu X, Cao Y, Yang F, Wang L, Li W, Xu Y, He L, Yu T, He G. Association study of 5-HT1A, 5-HT2A polymorphisms with schizophrenia and major depressive disorder in the Han Chinese population. Neurosci Lett 2016; 635:39-43. [PMID: 27756686 DOI: 10.1016/j.neulet.2016.10.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 10/03/2016] [Accepted: 10/11/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Rui Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China; Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China
| | - Yan Bi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China; Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China
| | - Weibo Niu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China; Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China
| | - Xiaoye Huang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China; Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China
| | - Shiqing Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China; Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China
| | - Xingwang Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China; Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China
| | - Xi Wu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China; Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China
| | - Yanfei Cao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China; Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China
| | - Fengping Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China; Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China
| | - Lu Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China; Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China
| | - Weidong Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China; Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China
| | - Yifeng Xu
- Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China; Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China
| | - Tao Yu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China; Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China.
| | - Guang He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China; Shanghai Key Laboratory of Psychiatric Disorders, Shanghai Mental Health Center, 600 South Wan Ping Road, Shanghai 200030, China.
| |
Collapse
|
30
|
Del Bello F, Cilia A, Carrieri A, Fasano DC, Ghelardini C, Di Cesare Mannelli L, Micheli L, Santini C, Diamanti E, Giannella M, Giorgioni G, Mammoli V, Paoletti CD, Petrelli R, Piergentili A, Quaglia W, Pigini M. The Versatile 2-Substituted Imidazoline Nucleus as a Structural Motif of Ligands Directed to the Serotonin 5-HT1A
Receptor. ChemMedChem 2016; 11:2287-2298. [DOI: 10.1002/cmdc.201600383] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 08/30/2016] [Indexed: 11/12/2022]
Affiliation(s)
- Fabio Del Bello
- School of Pharmacy; Medicinal Chemistry Unit; University of Camerino; Via S. Agostino 1 62032 Camerino Italy
| | - Antonio Cilia
- Recordati S.p.A.; Drug Discovery; via Civitali 1 20148 Milano (Italy)
| | - Antonio Carrieri
- Department of Pharmacy-Drug Science; University of Bari “Aldo Moro”; Via E. Orabona 4 70125 Bari Italy
| | - Domenico Claudio Fasano
- Department of Pharmacy-Drug Science; University of Bari “Aldo Moro”; Via E. Orabona 4 70125 Bari Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and; Child Health - Neurofarba - Pharmacology and Toxicology Section; University of Florence; Viale Pieraccini 6 50039 Firenze Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and; Child Health - Neurofarba - Pharmacology and Toxicology Section; University of Florence; Viale Pieraccini 6 50039 Firenze Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and; Child Health - Neurofarba - Pharmacology and Toxicology Section; University of Florence; Viale Pieraccini 6 50039 Firenze Italy
| | - Carlo Santini
- School of Science and Technology; University of Camerino; Via S. Agostino 1 62032 Camerino Italy
| | - Eleonora Diamanti
- School of Pharmacy; Medicinal Chemistry Unit; University of Camerino; Via S. Agostino 1 62032 Camerino Italy
| | - Mario Giannella
- School of Pharmacy; Medicinal Chemistry Unit; University of Camerino; Via S. Agostino 1 62032 Camerino Italy
| | - Gianfabio Giorgioni
- School of Pharmacy; Medicinal Chemistry Unit; University of Camerino; Via S. Agostino 1 62032 Camerino Italy
| | - Valerio Mammoli
- School of Pharmacy; Medicinal Chemistry Unit; University of Camerino; Via S. Agostino 1 62032 Camerino Italy
| | - Corinne Dalila Paoletti
- School of Pharmacy; Medicinal Chemistry Unit; University of Camerino; Via S. Agostino 1 62032 Camerino Italy
| | - Riccardo Petrelli
- School of Pharmacy; Medicinal Chemistry Unit; University of Camerino; Via S. Agostino 1 62032 Camerino Italy
| | - Alessandro Piergentili
- School of Pharmacy; Medicinal Chemistry Unit; University of Camerino; Via S. Agostino 1 62032 Camerino Italy
| | - Wilma Quaglia
- School of Pharmacy; Medicinal Chemistry Unit; University of Camerino; Via S. Agostino 1 62032 Camerino Italy
| | - Maria Pigini
- School of Pharmacy; Medicinal Chemistry Unit; University of Camerino; Via S. Agostino 1 62032 Camerino Italy
| |
Collapse
|
31
|
Sun N, Tischfield JA, King RA, Heiman GA. Functional Evaluations of Genes Disrupted in Patients with Tourette's Disorder. Front Psychiatry 2016; 7:11. [PMID: 26903887 PMCID: PMC4746269 DOI: 10.3389/fpsyt.2016.00011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 01/18/2016] [Indexed: 01/04/2023] Open
Abstract
Tourette's disorder (TD) is a highly heritable neurodevelopmental disorder with complex genetic architecture and unclear neuropathology. Disruptions of particular genes have been identified in subsets of TD patients. However, none of the findings have been replicated, probably due to the complex and heterogeneous genetic architecture of TD that involves both common and rare variants. To understand the etiology of TD, functional analyses are required to characterize the molecular and cellular consequences caused by mutations in candidate genes. Such molecular and cellular alterations may converge into common biological pathways underlying the heterogeneous genetic etiology of TD patients. Herein, we review specific genes implicated in TD etiology, discuss the functions of these genes in the mammalian central nervous system and the corresponding behavioral anomalies exhibited in animal models, and importantly, review functional analyses that can be performed to evaluate the role(s) that the genetic disruptions might play in TD. Specifically, the functional assays include novel cell culture systems, genome editing techniques, bioinformatics approaches, transcriptomic analyses, and genetically modified animal models applied or developed to study genes associated with TD or with other neurodevelopmental and neuropsychiatric disorders. By describing methods used to study diseases with genetic architecture similar to TD, we hope to develop a systematic framework for investigating the etiology of TD and related disorders.
Collapse
Affiliation(s)
- Nawei Sun
- Department of Genetics, Rutgers University, Piscataway, NJ, USA; Human Genetics Institute of New Jersey, Piscataway, NJ, USA
| | - Jay A Tischfield
- Department of Genetics, Rutgers University, Piscataway, NJ, USA; Human Genetics Institute of New Jersey, Piscataway, NJ, USA
| | - Robert A King
- Child Study Center, Yale School of Medicine , New Haven, CT , USA
| | - Gary A Heiman
- Department of Genetics, Rutgers University, Piscataway, NJ, USA; Human Genetics Institute of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
32
|
Goitia B, Rivero-Echeto MC, Weisstaub NV, Gingrich JA, Garcia-Rill E, Bisagno V, Urbano FJ. Modulation of GABA release from the thalamic reticular nucleus by cocaine and caffeine: role of serotonin receptors. J Neurochem 2015; 136:526-35. [PMID: 26484945 DOI: 10.1111/jnc.13398] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 09/23/2015] [Accepted: 10/02/2015] [Indexed: 11/30/2022]
Abstract
Serotonin receptors are targets of drug therapies for a variety of neuropsychiatric and neurodegenerative disorders. Cocaine inhibits the re-uptake of serotonin (5-HT), dopamine, and noradrenaline, whereas caffeine blocks adenosine receptors and opens ryanodine receptors in the endoplasmic reticulum. We studied how 5-HT and adenosine affected spontaneous GABAergic transmission from thalamic reticular nucleus. We combined whole-cell patch clamp recordings of miniature inhibitory post-synaptic currents (mIPSCs) in ventrobasal thalamic neurons during local (puff) application of 5-HT in wild type (WT) or knockout mice lacking 5-HT2A receptors (5-HT2A -/-). Inhibition of mIPSCs frequency by low (10 μM) and high (100 μM) 5-HT concentrations was observed in ventrobasal neurons from 5-HT2A -/- mice. In WT mice, only 100 μM 5-HT significantly reduced mIPSCs frequency. In 5-HT2A -/- mice, NAN-190, a specific 5-HT1A antagonist, prevented the 100 μM 5-HT inhibition while blocking H-currents that prolonged inhibition during post-puff periods. The inhibitory effects of 100 μM 5-HT were enhanced in cocaine binge-treated 5-HT2A -/- mice. Caffeine binge treatment did not affect 5-HT-mediated inhibition. Our findings suggest that both 5-HT1A and 5-HT2A receptors are present in pre-synaptic thalamic reticular nucleus terminals. Serotonergic-mediated inhibition of GABA release could underlie aberrant thalamocortical physiology described after repetitive consumption of cocaine. Our findings suggest that both 5-HT1A , 5-HT2A and A1 receptors are present in pre-synaptic TRN terminals. 5-HT1A and A1 receptors would down-regulate adenylate cyclase, whereas 5-HT1A would also increase the probability of the opening of G-protein-activated inwardly rectifying K(+) channels (GIRK). Sustained opening of GIRK channels would hyperpolarize pre-synaptic terminals activating H-currents, resulting in less GABA release. 5-HT2A -would activate PLC and IP3 , increasing intracellular [Ca(2+) ] and thus facilitating GABA release.
Collapse
Affiliation(s)
- Belén Goitia
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado" (DFBMC) Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET-UBA), Universidad de Buenos Aires, Ciudad Universitaria, Ciudad de Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - María Celeste Rivero-Echeto
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado" (DFBMC) Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET-UBA), Universidad de Buenos Aires, Ciudad Universitaria, Ciudad de Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Noelia V Weisstaub
- Grupo de Neurociencia de Sistemas, Departamento de Fisiología, Facultad de Medicina, Instituto de Fisiología y Biofísica (IFIBIO), UBA, Ciudad de Buenos Aires, Argentina
| | - Jay A Gingrich
- Division of Developmental Neuroscience, Columbia University and the NYSPI, Sackler Institute for Developmental Psychobiology, New York City, New York, USA
| | - Edgar Garcia-Rill
- Department of Neurobiology and Developmental Sciences, Center for Translational Neuroscience, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Verónica Bisagno
- Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Francisco J Urbano
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado" (DFBMC) Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET-UBA), Universidad de Buenos Aires, Ciudad Universitaria, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
33
|
Farb DH, Ratner MH. Targeting the modulation of neural circuitry for the treatment of anxiety disorders. Pharmacol Rev 2015; 66:1002-32. [PMID: 25237115 DOI: 10.1124/pr.114.009126] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Anxiety disorders are a major public health concern. Here, we examine the familiar area of anxiolysis in the context of a systems-level understanding that will hopefully lead to revealing an underlying pharmacological connectome. The introduction of benzodiazepines nearly half a century ago markedly improved the treatment of anxiety disorders. These agents reduce anxiety rapidly by allosterically enhancing the postsynaptic actions of GABA at inhibitory type A GABA receptors but side effects limit their use in chronic anxiety disorders. Selective serotonin reuptake inhibitors and serotonin/norepinephrine reuptake inhibitors have emerged as an effective first-line alternative treatment of such anxiety disorders. However, many individuals are not responsive and side effects can be limiting. Research into a relatively new class of agents known as neurosteroids has revealed novel modulatory sites and mechanisms of action that are providing insights into the pathophysiology of certain anxiety disorders, potentially bridging the gap between the GABAergic and serotonergic circuits underlying anxiety. However, translating the pharmacological activity of compounds targeted to specific receptor subtypes in rodent models of anxiety to effective therapeutics in human anxiety has not been entirely successful. Since modulating any one of several broad classes of receptor targets can produce anxiolysis, we posit that a systems-level discovery platform combined with an individualized medicine approach based on noninvasive brain imaging would substantially advance the development of more effective therapeutics.
Collapse
Affiliation(s)
- David H Farb
- Laboratory of Molecular Neurobiology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| | - Marcia H Ratner
- Laboratory of Molecular Neurobiology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
34
|
Viggiano A, Cacciola G, Widmer DAJ, Viggiano D. Anxiety as a neurodevelopmental disorder in a neuronal subpopulation: Evidence from gene expression data. Psychiatry Res 2015; 228:729-40. [PMID: 26089015 DOI: 10.1016/j.psychres.2015.05.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 05/14/2015] [Accepted: 05/26/2015] [Indexed: 12/20/2022]
Abstract
The relationship between genes and anxious behavior, is nor linear nor monotonic. To address this problem, we analyzed with a meta-analytic method the literature data of the behavior of knockout mice, retrieving 33 genes whose deletion was accompanied by increased anxious behavior, 34 genes related to decreased anxious behavior and 48 genes not involved in anxiety. We correlated the anxious behavior resulting from the deletion of these genes to their brain expression, using the Allen Brain Atlas and Gene Expression Omnibus (GEO) database. The main finding is that the genes accompanied, after deletion, by a modification of the anxious behavior, have lower expression in the cerebral cortex, the amygdala and the ventral striatum. The lower expression level was putatively due to their selective presence in a neuronal subpopulation. This difference was replicated also using a database of human gene expression, further showing that the differential expression pertained, in humans, a temporal window of young postnatal age (4 months up to 4 years) but was not evident at fetal or adult human stages. Finally, using gene enrichment analysis we also show that presynaptic genes are involved in the emergence of anxiety and postsynaptic genes in the reduction of anxiety after gene deletion.
Collapse
Affiliation(s)
- Adela Viggiano
- Department of Health Sciences, University of Molise, Campobasso 86100, Italy
| | - Giovanna Cacciola
- Department of Health Sciences, University of Molise, Campobasso 86100, Italy
| | | | - Davide Viggiano
- Department of Health Sciences, University of Molise, Campobasso 86100, Italy; Department of Cardio-Thoracic and Respiratory Science, Second University of Naples, Naples, Italy.
| |
Collapse
|
35
|
Stiedl O, Pappa E, Konradsson-Geuken Å, Ögren SO. The role of the serotonin receptor subtypes 5-HT1A and 5-HT7 and its interaction in emotional learning and memory. Front Pharmacol 2015; 6:162. [PMID: 26300776 PMCID: PMC4528280 DOI: 10.3389/fphar.2015.00162] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 07/20/2015] [Indexed: 11/13/2022] Open
Abstract
Serotonin [5-hydroxytryptamine (5-HT)] is a multifunctional neurotransmitter innervating cortical and limbic areas involved in cognition and emotional regulation. Dysregulation of serotonergic transmission is associated with emotional and cognitive deficits in psychiatric patients and animal models. Drugs targeting the 5-HT system are widely used to treat mood disorders and anxiety-like behaviors. Among the fourteen 5-HT receptor (5-HTR) subtypes, the 5-HT1AR and 5-HT7R are associated with the development of anxiety, depression and cognitive function linked to mechanisms of emotional learning and memory. In rodents fear conditioning and passive avoidance (PA) are associative learning paradigms to study emotional memory. This review assesses the role of 5-HT1AR and 5-HT7R as well as their interplay at the molecular, neurochemical and behavioral level. Activation of postsynaptic 5-HT1ARs impairs emotional memory through attenuation of neuronal activity, whereas presynaptic 5-HT1AR activation reduces 5-HT release and exerts pro-cognitive effects on PA retention. Antagonism of the 5-HT1AR facilitates memory retention possibly via 5-HT7R activation and evidence is provided that 5HT7R can facilitate emotional memory upon reduced 5-HT1AR transmission. These findings highlight the differential role of these 5-HTRs in cognitive/emotional domains of behavior. Moreover, the results indicate that tonic and phasic 5-HT release can exert different and potentially opposing effects on emotional memory, depending on the states of 5-HT1ARs and 5-HT7Rs and their interaction. Consequently, individual differences due to genetic and/or epigenetic mechanisms play an essential role for the responsiveness to drug treatment, e.g., by SSRIs which increase intrasynaptic 5-HT levels thereby activating multiple pre- and postsynaptic 5-HTR subtypes.
Collapse
Affiliation(s)
- Oliver Stiedl
- Department of Functional Genomics, Behavioral Neuroscience Group, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam - VU University Amsterdam Amsterdam, Netherlands ; Department of Molecular and Cellular Neurobiology, Behavioral Neuroscience Group, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam -VU University Amsterdam Amsterdam, Netherlands
| | - Elpiniki Pappa
- Department of Functional Genomics, Behavioral Neuroscience Group, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam - VU University Amsterdam Amsterdam, Netherlands ; Department of Molecular and Cellular Neurobiology, Behavioral Neuroscience Group, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam -VU University Amsterdam Amsterdam, Netherlands
| | | | - Sven Ove Ögren
- Department of Neuroscience, Karolinska Institutet Stockholm, Sweden
| |
Collapse
|
36
|
Effects of intra-infralimbic prefrontal cortex injections of cannabidiol in the modulation of emotional behaviors in rats: Contribution of 5HT1A receptors and stressful experiences. Behav Brain Res 2015; 286:49-56. [DOI: 10.1016/j.bbr.2015.02.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/06/2015] [Accepted: 02/10/2015] [Indexed: 01/27/2023]
|
37
|
Revisiting the Serotonin Hypothesis: Implications for Major Depressive Disorders. Mol Neurobiol 2015; 53:2778-2786. [PMID: 25823514 DOI: 10.1007/s12035-015-9152-z] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/19/2015] [Indexed: 02/06/2023]
Abstract
Major depressive disorder (MDD) is a heritable neuropsychiatric disease associated with severe changes at cellular and molecular levels. Its diagnosis mainly relies on the characterization of a wide range of symptoms including changes in mood and behavior. Despite the availability of antidepressant drugs, 10 to 30 % of patients fail to respond after a single or multiple treatments, and the recurrence of depression among responsive patients is very high. Evidence from the past decades suggests that the brain neurotransmitter serotonin (5-HT) is incriminated in MDD, and that a dysfunction of 5-HT receptors may play a role in the genesis of this disease. The 5-HT membrane transporter protein (SERT), which helps regulate the serotonergic transmission, is also implicated in MDD and is one of the main targets of antidepressant therapy. Although a number of behavioral tests and animal models have been developed to study depression, little is known about the neurobiological bases of MDD. Understanding the role of the serotonergic pathway will significantly help improve our knowledge of the pathophysiology of depression and may open up avenues for the development of new antidepressant drugs. The overarching goal of this review is to present recent findings from studies examining the serotonergic pathway in MDD, with a focus on SERT and the serotonin 1A (5-HT1A), serotonin 1B (5-HT1B), and serotonin 2A (5-HT2A) receptors. This paper also describes some of the main molecules involved in the internalization of 5-HT receptors and illustrates the changes in 5-HT neurotransmission in knockout mice and animal model of depression.
Collapse
|
38
|
Piszczek L, Piszczek A, Kuczmanska J, Audero E, Gross CT. Modulation of anxiety by cortical serotonin 1A receptors. Front Behav Neurosci 2015; 9:48. [PMID: 25759645 PMCID: PMC4338812 DOI: 10.3389/fnbeh.2015.00048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 02/09/2015] [Indexed: 01/29/2023] Open
Abstract
Serotonin (5-HT) plays an important role in the modulation of behavior across animal species. The serotonin 1A receptor (Htr1a) is an inhibitory G-protein coupled receptor that is expressed both on serotonin and non-serotonin neurons in mammals. Mice lacking Htr1a show increased anxiety behavior suggesting that its activation by serotonin has an anxiolytic effect. This outcome can be mediated by either Htr1a population present on serotonin (auto-receptor) or non-serotonin neurons (hetero-receptor), or both. In addition, both transgenic and pharmacological studies have shown that serotonin acts on Htr1a during development to modulate anxiety in adulthood, demonstrating a function for this receptor in the maturation of anxiety circuits in the brain. However, previous studies have been equivocal about which Htr1a population modulates anxiety behavior, with some studies showing a role of Htr1a hetero-receptor and others implicating the auto-receptor. In particular, cell-type specific rescue and suppression of Htr1a expression in either forebrain principal neurons or brainstem serotonin neurons reached opposite conclusions about the role of the two populations in the anxiety phenotype of the knockout. One interpretation of these apparently contradictory findings is that the modulating role of these two populations depends on each other. Here we use a novel Cre-dependent inducible allele of Htr1a in mice to show that expression of Htr1a in cortical principal neurons is sufficient to modulate anxiety. Together with previous findings, these results support a hetero/auto-receptor interaction model for Htr1a function in anxiety.
Collapse
Affiliation(s)
- Lukasz Piszczek
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| | - Agnieszka Piszczek
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| | - Joanna Kuczmanska
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| | - Enrica Audero
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| | - Cornelius T Gross
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| |
Collapse
|
39
|
5-HT1A receptor-dependent control of nigrostriatal dopamine neurotransmission in the pharmacotherapy of Parkinson’s disease and schizophrenia. Behav Pharmacol 2015; 26:45-58. [DOI: 10.1097/fbp.0000000000000123] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
40
|
Luscher B, Fuchs T. GABAergic control of depression-related brain states. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2015; 73:97-144. [PMID: 25637439 DOI: 10.1016/bs.apha.2014.11.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The GABAergic deficit hypothesis of major depressive disorders (MDDs) posits that reduced γ-aminobutyric acid (GABA) concentration in brain, impaired function of GABAergic interneurons, altered expression and function of GABA(A) receptors, and changes in GABAergic transmission dictated by altered chloride homeostasis can contribute to the etiology of MDD. Conversely, the hypothesis posits that the efficacy of currently used antidepressants is determined by their ability to enhance GABAergic neurotransmission. We here provide an update for corresponding evidence from studies of patients and preclinical animal models of depression. In addition, we propose an explanation for the continued lack of genetic evidence that explains the considerable heritability of MDD. Lastly, we discuss how alterations in GABAergic transmission are integral to other hypotheses of MDD that emphasize (i) the role of monoaminergic deficits, (ii) stress-based etiologies, (iii) neurotrophic deficits, and (iv) the neurotoxic and neural circuit-impairing consequences of chronic excesses of glutamate. We propose that altered GABAergic transmission serves as a common denominator of MDD that can account for all these other hypotheses and that plays a causal and common role in diverse mechanistic etiologies of depressive brain states and in the mechanism of action of current antidepressant drug therapies.
Collapse
Affiliation(s)
- Bernhard Luscher
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, USA; Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, USA; Center for Molecular Investigation of Neurological Disorders, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA.
| | - Thomas Fuchs
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, USA; Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, USA; Center for Molecular Investigation of Neurological Disorders, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
41
|
Seven A, Yüksel B, Kılıç S, Esen H, Keskin U, Ulubay M, Ozekinci M. Effect of injectable medroxyprogesterone acetate and etonogestrel implants on GABA-A and serotonin receptors in white and gray matter of the brain: experimental study in rats. Gynecol Endocrinol 2014; 30:320-4. [PMID: 24460500 DOI: 10.3109/09513590.2014.880417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The aim of this study was to evaluate the time-dependent effect of progesterone-only contraceptives on the brain and to obtain an improved understanding of mood disorders experienced under this medication. A total of 66 Wistar albino rats were divided into three groups: etonogestrel (ENG) implant (group 1, n = 30); depot medroxyprogesterone acetate (MPA)-injectable (group 2, n = 30); and control (group 3, n = 6) groups. Groups 1 and 2 were each divided into five subgroups, which were examined every 10 d for up to 50 d after medication administration, to evaluate its time-dependent effect. There was no difference in terms of gamma-aminobutyric acid (GABA) and serotonin immunohistochemical staining in white and gray matter among the subgroups of group 1. In group 2, there was a significant decrease in serotonin receptor staining intensity in white and gray matter on day 50, when compared to the control group (p = 0.041). When the subgroups of group 2 were compared, there was a significant decrease in serotonin receptor staining intensity in white and gray matter on days 40 and 50 when compared to day 10. In conclusion, we showed that ENG and MPA have no effect on apoptosis and GABA-A receptors in the brain. We also showed that MPA has time-dependent effects on serotonin receptors, which may be a possible mechanism involved in mood disorders during long-term usage of injectable progesterone-only contraceptives.
Collapse
Affiliation(s)
- Ali Seven
- Department of Obstetrics and Gynaecology, Dumlupinar University, Faculty of Medicine , Kutahya , Turkey
| | | | | | | | | | | | | |
Collapse
|
42
|
Kim MH, Leem YH. Chronic exercise improves repeated restraint stress-induced anxiety and depression through 5HT1A receptor and cAMP signaling in hippocampus. J Exerc Nutrition Biochem 2014; 18:97-104. [PMID: 25566444 PMCID: PMC4241932 DOI: 10.5717/jenb.2014.18.1.97] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 02/05/2014] [Accepted: 02/26/2014] [Indexed: 11/11/2022] Open
Abstract
[Purpose] Mood disorders such as anxiety and depression are prevalent psychiatric illness, but the role of 5HT1A in the anti-depressive effects of exercise has been rarely known yet. We investigated whether long-term exercise affected a depressive-like behavior and a hippocampal 5HT1A receptor-mediated cAMP/PKA/CREB signaling in depression mice model. [Methods] To induce depressive behaviors, mice were subjected to 14 consecutive days of restraint stress (2 hours/day). Depression-like behaviors were measured by forced swimming test (TST), and anxiety-like behavior was assessed by elevated plus maze (EPM). Treadmill exercise was performed with 19 m/min for 60 min/day, 5 days/week from weeks 0 to 8. Restraint stress was started at week 6 week and ended at week 8. To elucidate the role of 5HT1A in depression, the immunoreactivities of 5HT1A were detected in hippocampus using immunohistochemical technique. [Results] Chronic/repeated restraint stress induced behavioral anxiety and depression, such as reduced time and entries in open arms in EPM and enhanced immobility time in FST. These anxiety and depressive behaviors were ameliorated by chronic exercise. Also, these behavioral changes were concurrent with the deficit of 5HT1A and cAMP/PKA/CREB cascade in hippocampus, which was coped with chronic exercise. [Conclusion] These results suggest that chronic exercise may improve the disturbance of hippocampal 5HT1A-regulated cAMP/PKA/CREB signaling in a depressed brain, thereby exerting an antidepressive action.
Collapse
Affiliation(s)
- Mun Hee Kim
- Exercise Biochemistry Laboratory, Korea National Sport University, Seoul, Korea
| | - Yea Hyun Leem
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan-si, Korea
| |
Collapse
|
43
|
Maximino C, Puty B, Benzecry R, Araújo J, Lima MG, de Jesus Oliveira Batista E, Renata de Matos Oliveira K, Crespo-Lopez ME, Herculano AM. Role of serotonin in zebrafish (Danio rerio) anxiety: Relationship with serotonin levels and effect of buspirone, WAY 100635, SB 224289, fluoxetine and para-chlorophenylalanine (pCPA) in two behavioral models. Neuropharmacology 2013; 71:83-97. [DOI: 10.1016/j.neuropharm.2013.03.006] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 03/04/2013] [Accepted: 03/12/2013] [Indexed: 12/21/2022]
|
44
|
Razoux F, Baltes C, Mueggler T, Seuwen A, Russig H, Mansuy I, Rudin M. Functional MRI to assess alterations of functional networks in response to pharmacological or genetic manipulations of the serotonergic system in mice. Neuroimage 2013; 74:326-36. [DOI: 10.1016/j.neuroimage.2013.02.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 01/17/2013] [Accepted: 02/11/2013] [Indexed: 01/21/2023] Open
|
45
|
Li M, Xue X, Shao S, Shao F, Wang W. Cognitive, emotional and neurochemical effects of repeated maternal separation in adolescent rats. Brain Res 2013; 1518:82-90. [PMID: 23623774 DOI: 10.1016/j.brainres.2013.04.026] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 04/12/2013] [Accepted: 04/12/2013] [Indexed: 02/06/2023]
Abstract
As an adverse early life experience, maternal separation (MS) induces profound neurochemical, cognitive and emotional dysfunction. Previous studies have reported that MS affected prepulse inhibition (PPI), anxiety-related behaviors, dopaminergic and serotonergic activity in adult rats, and in the present study, we investigated the effects of repeated (4h/day) maternal separation during postnatal days 1-21 on PPI and anxiety-related behaviors in an elevated plus maze, as well as dopamine D2 receptor (DRD2) and 5-HT1A receptor expression in the medial prefrontal cortex (mPFC), nucleus accumbens (NAc) and hippocampus in adolescent rats. Our findings show that repeated MS results in reduced PPI, increased anxiety-related behaviors, decreased DRD2 protein expression in the NAc and hippocampus, and decreased 5-HT1A protein expression in the mPFC and hippocampus in adolescent rats. These data further demonstrate that MS can be used as an animal model of neuropsychiatric disease.
Collapse
Affiliation(s)
- Man Li
- Department of Psychology, Peking University, 5 Yiheyuan Road, Beijing 100871, China
| | | | | | | | | |
Collapse
|
46
|
Kaushik M, Lamberton PHL, Webster JP. The role of parasites and pathogens in influencing generalised anxiety and predation-related fear in the mammalian central nervous system. Horm Behav 2012; 62:191-201. [PMID: 22521209 DOI: 10.1016/j.yhbeh.2012.04.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 03/30/2012] [Accepted: 04/04/2012] [Indexed: 01/15/2023]
Abstract
Behavioural and neurophysiological traits and responses associated with anxiety and predation-related fear have been well documented in rodent models. Certain parasites and pathogens which rely on predation for transmission appear able to manipulate these, often innate, traits to increase the likelihood of their life-cycle being completed. This can occur through a range of mechanisms, such as alteration of hormonal and neurotransmitter communication and/or direct interference with the neurons and brain regions that mediate behavioural expression. Whilst some post-infection behavioural changes may reflect 'general sickness' or a pathological by-product of infection, others may have a specific adaptive advantage to the parasite and be indicative of active manipulation of host behaviour. Here we review the key mechanisms by which anxiety and predation-related fears are controlled in mammals, before exploring evidence for how some infectious agents may manipulate these mechanisms. The protozoan Toxoplasma gondii, the causative agent of toxoplasmosis, is focused on as a prime example. Selective pressures appear to have allowed this parasite to evolve strategies to alter the behaviour in its natural intermediate rodent host. Latent infection has also been associated with a range of altered behavioural profiles, from subtle to severe, in other secondary host species including humans. In addition to enhancing our knowledge of the evolution of parasite manipulation in general, to further our understanding of how and when these potential changes to human host behaviour occur, and how we may prevent or manage them, it is imperative to elucidate the associated mechanisms involved.
Collapse
Affiliation(s)
- Maya Kaushik
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College Faculty of Medicine, St Mary's Hospital Campus, Norfolk Place, London W2 1PG, UK
| | | | | |
Collapse
|
47
|
Huot P, Johnston TH, Visanji NP, Darr T, Pires D, Hazrati LN, Brotchie JM, Fox SH. Increased levels of 5-HT1A receptor binding in ventral visual pathways in Parkinson's disease. Mov Disord 2012; 27:735-42. [PMID: 22419526 DOI: 10.1002/mds.24964] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 01/27/2012] [Accepted: 02/09/2012] [Indexed: 11/09/2022] Open
Abstract
Visual hallucinations are common in advanced Parkinson's disease (PD). The pathophysiology of visual hallucinations may involve enhanced serotonergic neurotransmission. The atypical antipsychotics clozapine and quetiapine, which have affinity for 5-HT(2A) and 5-HT(1A) receptors, are effective against visual hallucinations in PD. 5-HT(2A) receptors are increased in ventral visual pathways in PD patients with visual hallucinations, and we hypothesized that 5-HT(1A) receptors were also involved in visual hallucinations in PD. Autoradiographic binding using [(3) H]-WAY-100,635 and NAN-190 was performed in brain sections from 6 PD patients with visual hallucinations, 6 PD patients without visual hallucinations, and 5 age-matched controls. All PD subjects had been treated with L-dopa. Brain areas studied were the orbitofrontal, inferolateral temporal, and motor cortices, as well as the striatum, globus pallidus, substantia nigra, and thalamus. 5-HT(1A) -binding levels were dramatically increased in the ventral visual pathways of all PD patients compared with controls (0 vs 11 and 0 vs 100 nmol/mg, respectively; both P < .05). There was no significant difference in 5-HT(1A) -binding levels in PD patients with visual hallucinations compared with PD patients without visual hallucinations or with controls in any of the brain areas studied (P > .05). Gross abnormalities in 5-HT(1A) levels in ventral visual areas occurred in all PD patients exposed to L-dopa. However, as there was no difference in 5-HT(1A) -binding levels between hallucinators and nonhallucinators, alterations in 5-HT(1A) receptor levels may not contribute specifically to visual hallucinations in PD. However, the discrete anatomical distribution of rises to the ventral visual areas suggests some role in predisposing to visual hallucinations.
Collapse
Affiliation(s)
- Philippe Huot
- Toronto Western Research Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Li Q, Luo T, Jiang X, Wang J. Anxiolytic effects of 5-HT₁A receptors and anxiogenic effects of 5-HT₂C receptors in the amygdala of mice. Neuropharmacology 2011; 62:474-84. [PMID: 21925519 DOI: 10.1016/j.neuropharm.2011.09.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 09/02/2011] [Accepted: 09/03/2011] [Indexed: 01/14/2023]
Abstract
The aim of the present study is to test a hypothesis that 5-HT(1A) and 5-HT(2C) receptors in the amygdala play an important role in the regulation of anxiety behaviors. We examined alterations in anxiety-like behaviors after manipulation of the expression of 5-HT(1A) and 5-HT(2C) receptors in the amygdala using recombinant adenovirus approaches. Recombinant adenoviruses containing a 5-HT(1A) promoter-controlled 5-HT(1A) antisense sequence or a 5-HT(2C) promoter-controlled 5-HT(2C) sense sequence were injected into the amygdala. Elevated plus-maze (EPM) and open field tests were conducted to determine anxiety-like behavior and locomotor activity. Reductions in the expression of 5-HT(1A) receptors in the amygdala significantly attenuated the time spent in the open arms of EPM and time spent in the center of an open field. Reduction in the percent of time spent in the open arms of EPM is negatively correlated with the density of 5-HT(1A) receptors in the central amygdala. On the other hand, increased expression of 5-HT(2C) receptors reduced the time spent in the open arms of EPM and time spent in the center of an open field. The reductions in the time spent and distance traveled in the open arms of EPM were correlated to the density of 5-HT(2C) receptors in the basolateral nucleus of amygdala. These data suggest that amygdaloid 5-HT(1A) receptors produce anxiolytic and 5-HT(2C) receptors produce anxiogenic effects. Together, the present results demonstrate the important role of the amygdaloid 5-HT(1A) and 5-HT(2C) receptors in the regulation of anxiety-like behaviors. This article is part of a Special Issue entitled 'Anxiety and Depression'.
Collapse
Affiliation(s)
- Qian Li
- Department of Psychiatry and Behavioral Sciences, University of Texas Medical Branch, Galveston, TX, USA.
| | | | | | | |
Collapse
|
49
|
Characterization of 5-HT1A/1B−/− mice: An animal model sensitive to anxiolytic treatments. Neuropharmacology 2011; 61:478-88. [DOI: 10.1016/j.neuropharm.2011.02.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 02/07/2011] [Accepted: 02/09/2011] [Indexed: 11/24/2022]
|
50
|
Huot P, Fox SH, Brotchie JM. The serotonergic system in Parkinson's disease. Prog Neurobiol 2011; 95:163-212. [PMID: 21878363 DOI: 10.1016/j.pneurobio.2011.08.004] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 08/05/2011] [Accepted: 08/15/2011] [Indexed: 01/23/2023]
Abstract
Although the cardinal manifestations of Parkinson's disease (PD) are attributed to a decline in dopamine levels in the striatum, a breadth of non-motor features and treatment-related complications in which the serotonergic system plays a pivotal role are increasingly recognised. Serotonin (5-HT)-mediated neurotransmission is altered in PD and the roles of the different 5-HT receptor subtypes in disease manifestations have been investigated. The aims of this article are to summarise and discuss all published preclinical and clinical studies that have investigated the serotonergic system in PD and related animal models, in order to recapitulate the state of the current knowledge and to identify areas that need further research and understanding.
Collapse
Affiliation(s)
- Philippe Huot
- Toronto Western Research Institute, MCL 11-419, Toronto Western Hospital, University Health Network, 399 Bathurst Street, Toronto, Ontario, Canada M5T 2S8
| | | | | |
Collapse
|