1
|
Zhao L, Witter MP, Palomero-Gallagher N. Cyto-, gene, and multireceptor architecture of the early postnatal mouse hippocampal complex. Prog Neurobiol 2024; 245:102704. [PMID: 39709019 DOI: 10.1016/j.pneurobio.2024.102704] [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: 10/13/2024] [Revised: 11/27/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Neurotransmitter receptors are key molecules in signal transmission in the adult brain, and their precise spatial and temporal balance expressions also play a critical role in normal brain development. However, the specific balance expression of multiple receptors during hippocampal development is not well characterized. In this study, we used quantitative in vivo receptor autoradiography to measure the distributions and densities of 18 neurotransmitter receptor types in the mouse hippocampal complex at postnatal day 7, and compared them with the expressions of their corresponding encoding genes. We provide a novel and comprehensive characterization of the cyto-, gene, and multireceptor architecture of the developing mouse hippocampal and subicular regions during the developmental period, which typically differs from that in the adult brain. High-density receptor expressions with distinct regional and laminar distributions were observed for AMPA, Kainate, mGluR2/3, GABAA, GABAA/BZ, α2, and A1 receptors during this specific period, whereas NMDA, GABAB, α1, M1, M2, M3, nicotinic α4β2, 5-HT1A, 5-HT2, D1 and D2/D3 receptors exhibited relatively low and homogeneous expressions. This specific balance of multiple receptors aligns with regional cytoarchitecture, neurotransmitter distributions, and gene expressions. Moreover, contrasting with previous findings, we detected a high α2 receptor density, with distinct regional and laminar distribution patterns. A non-covariation differentiation phenomenon between α2 receptor distributions and corresponding gene expressions is also demonstrated in this early developmental period. The multimodal data provides new insights into understanding the hippocampal development from the perspective of cell, gene, and multireceptor levels, and contributes important resources for further interdisciplinary analyses.
Collapse
Affiliation(s)
- Ling Zhao
- Department of Psychology, School of Public Policy and Management, Nanchang University, Nanchang 330000, China; Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich 52425, Germany.
| | - Menno P Witter
- Kavli Institute for Systems Neuroscience, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Nicola Palomero-Gallagher
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich 52425, Germany; C. & O. Vogt Institute for Brain Research, Heinrich-Heine-University, Dusseldorf 40225, Germany
| |
Collapse
|
2
|
Valipour H, Meftahi GH, Pirzad Jahromi G, Mohammadi A. Lateralization of the 5-HT 1A receptors in the basolateral amygdala in metabolic and anxiety responses to chronic restraint stress. Amino Acids 2024; 56:13. [PMID: 38340185 PMCID: PMC10858818 DOI: 10.1007/s00726-023-03380-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 12/24/2023] [Indexed: 02/12/2024]
Abstract
Behavioral and functional studies describe hemispheric asymmetry in anxiety and metabolic behaviors in responses to stress. However, no study has reported serotonergic receptor (the 5-HT1A receptor) lateralization in the basolateral amygdala (BLA) in vivo on anxiety and metabolic behaviors under stress. In the present study, the effect of unilateral and bilateral suppression of the 5-HT1A receptor in the BLA on anxiety, and metabolic responses to chronic restraint stress was assessed. Male Wistar rats 7 days after cannulation into the BLA received chronic restraint stress for 14 consecutive days. 20 minutes before induction of stress, WAY-100-635 (selective 5-HT1A antagonist) or sterile saline (vehicle) was administered either uni- or bi-laterally into the BLA. Behavioral (elevated plus maze; EPM, and open field test), and metabolic parameter studies were performed. Results showed that stress causes a significant increase in weight gain compared to control. In the non-stress condition, the left and bilaterally, and in the stress condition the right, left, and both sides, inhibition of 5-HT1A in the BLA reduced weight gain. In the restraint stress condition, only inhibition of the 5-HT1A receptor in the left BLA led to decreased food intake compared to the control group. In stress conditions, inhibition of the 5-HT1A receptor on the right, left, and bilateral BLA increased water intake compared to the stress group. Inhibition of the 5-HT1A receptor on the left side of the BLA by WAY-100-635 induced anxiety-like behaviors in stressed rats. Similarly, WAY-100-635 on the left BLA effectively caused anxiety-like behaviors in both EPM and open field tests in the control animals. In conclusion, it seems that 5-HT1A receptors in the left BLA are more responsible for anxiety-like behaviors and metabolic changes in responses to stress.
Collapse
Affiliation(s)
- Habib Valipour
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Gholam Hossein Meftahi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Gila Pirzad Jahromi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Mohammadi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Chda A, Bencheikh R. Flavonoids as G Protein-coupled Receptors Ligands: New Potential Therapeutic Natural Drugs. Curr Drug Targets 2023; 24:1346-1363. [PMID: 38037994 DOI: 10.2174/0113894501268871231127105219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023]
Abstract
G protein coupled receptors (GPCRs) are among the largest family of cell surface receptors found in the human genome. They govern a wide range of physiological responses in both health and diseases, making them one of the potential targeted surface receptors for pharmaceuticals. Flavonoids can modulate GPCRs activity by acting as allosteric ligands. They can either enhance or reduce the GPCR's effect. Emerging research shows that individual flavonoids or mixtures of flavonoids from plant extracts can have relevant pharmacological effects against a number of diseases, particularly by influencing GPCRs. In the present review, we are considering to give a comprehensive overview of flavonoids and related compounds that exhibit GPCRs activity and to further explore which beneficial structural features. Molecular docking was used to strengthen experimental evidence and describe flavonoid-GPCRs interactions at molecular level.
Collapse
Affiliation(s)
- Alae Chda
- Laboratory of Microbial Biotechnology and Bioactive Molecules (LM2BM), Faculty of Science and Technology, Sidi Mohamed Ben Abdellah University, Road of Immouzer, PO Box 2202, Fez, Morocco
- Higher Institute of Nursing and Health Techniques - Fez. Ministry of Health and Social Protection, Fez, Morocco
| | - Rachid Bencheikh
- Laboratory of Microbial Biotechnology and Bioactive Molecules (LM2BM), Faculty of Science and Technology, Sidi Mohamed Ben Abdellah University, Road of Immouzer, PO Box 2202, Fez, Morocco
| |
Collapse
|
4
|
Kyi-Tha-Thu C, Fujitani Y, Hirano S, Win-Shwe TT. Early-Life Exposure to Traffic-Related Air Pollutants Induced Anxiety-like Behaviors in Rats via Neurotransmitters and Neurotrophic Factors. Int J Mol Sci 2022; 24:ijms24010586. [PMID: 36614034 PMCID: PMC9820394 DOI: 10.3390/ijms24010586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/06/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Recent epidemiological studies have reported significantly increasing hospital admission rates for mental disorders such as anxiety and depression, not only in adults but also in children and adolescents, indicating more research is needed for evaluation of the etiology and possible reduction and prevention of these disorders. The aim of the present study was to examine the associations between perinatal exposure to traffic-related air pollutants and anxiety-like behaviors and alterations in neurological and immunological markers in adulthood using a rat model. Sprague Dawley pregnant rats were exposed to clean air (control), diesel exhaust (DE) 101 ± 9 μg/m3 or diesel exhaust origin secondary organic aerosol (DE-SOA) 118 ± 23 μg/m3 from gestational day 14 to postnatal day 21. Anxiety-related behavioral tests including open field tests, elevated plus maze, light/dark transition tests and novelty-induced hypophagia were performed on 10-week-old rats. The hippocampal expression of neurotransmitters, neurotrophic factors, and inflammatory molecular markers was examined by real-time RT-PCR. Anxiety-like behaviors were observed in both male and female rat offspring exposed to DE or DE-SOA. Moreover, serotonin receptor (5HT1A), dopamine receptor (Drd2), brain-derived neurotrophic factor and vascular endothelial growth factor A mRNAs were significantly decreased, whereas interleukin-1β, cyclooxygenase-2, heme oxygenase-1 mRNAs and microglial activation were significantly increased in both male and female rats. These findings indicate that brain developmental period exposure to traffic-related air pollutants may induce anxiety-like behaviors via modulation of neurotransmitters, neurotrophic factors, and immunological molecular markers, triggering neuroinflammation and microglia activation in rats.
Collapse
Affiliation(s)
- Chaw Kyi-Tha-Thu
- Department of Immunology, School of Medicine, International University of Health and Welfare, 4-3, Kozunomori, Narita 286-8686, Chiba, Japan
| | - Yuji Fujitani
- Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba 305-8506, Ibaraki, Japan
| | - Seishiro Hirano
- Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba 305-8506, Ibaraki, Japan
| | - Tin-Tin Win-Shwe
- Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba 305-8506, Ibaraki, Japan
- Correspondence: ; Tel.: +81-29-850-2542
| |
Collapse
|
5
|
Jalal K, Khan F, Nawaz S, Afroz R, Khan K, Ali SB, Hao L, Khan SA, Kazi M, Uddin R, Haleem DJ. Anxiolytic, anti-nociceptive and body weight reducing effects of L-lysine in rats: Relationship with brain serotonin an In-Vivo and In-Silico study. Biomed Pharmacother 2022; 152:113235. [PMID: 35696944 DOI: 10.1016/j.biopha.2022.113235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 11/17/2022] Open
Abstract
L-lysine (L-lys) had long been comprehended as an essential amino acid for humans. There were reports that the absence or inadequate availability of L-lys in the diet may lead to mental and physical impairments. The present study was designed to explore the effects of L-lys on body weight changes, cumulative food intake, anxiety-like behavior and pain perception in rats. 5-Hydroxytryptamine (5-HT, serotonin) metabolism, and tryptophan (Trp) levels in the midbrain (MB), hippocampus (HP), and prefrontal cortex (PFC) were also determined. Animals were treated with L-lys in doses of 0.5 g/kg and 1 g/kg for 20 days and behavioral studies were performed on day 1st and day 20th. After monitoring behaviors on day 20th, animals were killed to collect the serum and brain regions MB, HP and PFC. 5-HT metabolism and Trp levels were determined by HPLC-EC. The treatment produce no effect on food intakes but body weights were reduced. 20 days administration of L-lys produced an anxiolytic effect and increased exploratory activity on day 1st. Repeated administration of L-lys increased 5-HT levels in the PFC and HP. 5-Hydroxyindoleacetic acid (5-HIAA), the metabolite of 5-HT, decreased in the HP. Trp, the precourser of 5-HT, decreased in the PFC. Results suggested a decrease in 5-HT degredation in enhancing 5-HT levels. Results of in-silico analysis showed that lysine had a potential binding affinity for MAO (monoamine oxidase) A and B with an energy of (-4.8 kcal/mol and -5.3 kcal/mol) respectively. The molecular dynamic simulation study revealed the stability of L-lys after 10 ns for each protein. Conclusively, the present study showed that L-lys produced an anxiolytic effect and reduced body weight. These beneficial effects were associated with an increase in 5-HT levels in the PFC and HP. In-silico analysis suggested that 5-HT increase were due to the binding of L-lys with MAOs resulting in an inhibition of the degradation of monoamine.
Collapse
Affiliation(s)
- Khurshid Jalal
- HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| | - Faisal Khan
- HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Shazia Nawaz
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Rushda Afroz
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Kanwal Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Sadia Basharat Ali
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Liangliang Hao
- Hospital of Chengdu University of Traditional Chinese Medicine, No.39 Shi-er-qiao Road, Chengdu, PR China
| | - Saeed Ahmad Khan
- Department of Pharmacy, Kohat University of Science and Technology, KP, Pakistan; Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin,78712, USA
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, PO Box 2457, Riyadh 11451, Saudi Arabia
| | - Reaz Uddin
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Darakhshan Jabeen Haleem
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
6
|
Chronic hM4Di-DREADD-Mediated Chemogenetic Inhibition of Forebrain Excitatory Neurons in Postnatal or Juvenile Life Does Not Alter Adult Mood-Related Behavior. eNeuro 2022; 9:ENEURO.0381-21.2021. [PMID: 35115382 PMCID: PMC8856708 DOI: 10.1523/eneuro.0381-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/21/2021] [Accepted: 12/25/2021] [Indexed: 11/21/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) coupled to Gi signaling, in particular downstream of monoaminergic neurotransmission, are posited to play a key role during developmental epochs (postnatal and juvenile) in shaping the emergence of adult anxiodepressive behaviors and sensorimotor gating. To address the role of Gi signaling in these developmental windows, we used a CaMKIIα-tTA::TRE hM4Di bigenic mouse line to express the hM4Di-DREADD (designer receptor exclusively activated by designer drugs) in forebrain excitatory neurons and enhanced Gi signaling via chronic administration of the DREADD agonist, clozapine-N-oxide (CNO) in the postnatal window (postnatal days 2–14) or the juvenile window (postnatal days 28–40). We confirmed that the expression of the HA-tagged hM4Di-DREADD was restricted to CaMKIIα-positive neurons in the forebrain, and that the administration of CNO in postnatal or juvenile windows evoked inhibition in forebrain circuits of the hippocampus and cortex, as indicated by a decline in expression of the neuronal activity marker c-Fos. hM4Di-DREADD-mediated inhibition of CaMKIIα-positive forebrain excitatory neurons in postnatal or juvenile life did not impact the weight profile of mouse pups, and also did not influence the normal ontogeny of sensory reflexes. Further, postnatal or juvenile hM4Di-DREADD-mediated inhibition of CaMKIIα-positive forebrain excitatory neurons did not alter anxiety- or despair-like behaviors in adulthood and did not impact sensorimotor gating. Collectively, these results indicate that chemogenetic induction of Gi signaling in CaMKIIα-positive forebrain excitatory neurons in postnatal and juvenile temporal windows does not appear to impinge on the programming of anxiodepressive behaviors in adulthood.
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
|
Trujillo V, Valentim-Lima E, Mencalha R, Carbalan QSR, Dos-Santos RC, Felintro V, Girardi CEN, Rorato R, Lustrino D, Reis LC, Mecawi AS. Neonatal Serotonin Depletion Induces Hyperactivity and Anxiolytic-like Sex-Dependent Effects in Adult Rats. Mol Neurobiol 2020; 58:1036-1051. [PMID: 33083963 DOI: 10.1007/s12035-020-02181-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/14/2020] [Indexed: 12/16/2022]
Abstract
The serotoninergic system plays an important role in the ontogeny of the mammalian central nervous system, and changes in serotonin production during development may lead to permanent changes in brain cytoarchitecture and function. The present study investigated the programming effects of neonatal serotonin depletion on behavior and molecular components of the serotoninergic system in adult male and female rats. Subcutaneous para-chlorophenylalanine (pCPA) administration (100 mg kg-1) was performed daily on postnatal days 8-16 to deplete brain serotonin content. During adulthood, elevated plus-maze, open field, social interaction, forced swimming, and food, saline, and sucrose intake tests were performed. Relative expression of serotonin neurotransmission components in several brain areas was determined by qPCR. Additionally, serotonin immunofluorescence and neuropeptide mRNA expression were assessed in dorsal raphe (DRN) and paraventricular (PVN) nuclei, respectively. Rat performance in behavioral tests demonstrated a general increase in locomotor activity and active escape behavior as well as decreased anxiety-like behavior after neonatal brain serotonin depletion. The behavioral programming effects due to neonatal serotonin depletion were more pronounced in females than males. At the gene expression level, the mRNA of Tph1 and Tph2 were lower in DRN while Htr2c was higher in the amygdala of pCPA-treated males, while Htr1a, Htr2c, Oxt, Avp, Crh, and Trh were not different in any treatments or sex in PVN. The results indicate that neonatal serotonin depletion has long-term consequences on locomotion and anxiety-like behavior associated with long-lasting molecular changes in the brain serotoninergic system in adult rats.
Collapse
Affiliation(s)
- Verónica Trujillo
- Laboratory of Neuroendocrinology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 862, Edifício de Ciências Biomédicas, 7° andar, Vila Clementino, São Paulo, CEP 04023-062, Brasil
- Departament of Physiology, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Evandro Valentim-Lima
- Laboratory of Neuroendocrinology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 862, Edifício de Ciências Biomédicas, 7° andar, Vila Clementino, São Paulo, CEP 04023-062, Brasil
| | - Rodrigo Mencalha
- Department of Natural Sciences, Universidade Federal do Acre, Rio Branco, Brazil
| | - Quézia S R Carbalan
- Department of Physiological Sciences, Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil
| | - Raoni C Dos-Santos
- Department of Physiological Sciences, Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil
| | - Viviane Felintro
- Department of Physiological Sciences, Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil
| | - Carlos E N Girardi
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Rodrigo Rorato
- Laboratory of Neuroendocrinology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 862, Edifício de Ciências Biomédicas, 7° andar, Vila Clementino, São Paulo, CEP 04023-062, Brasil
| | - Danilo Lustrino
- Department of Physiology, Centro de Ciências Biológicas e da Saúde, Universidade Federal de Sergipe, São Cristóvão, Brazil
| | - Luis C Reis
- Department of Physiological Sciences, Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil
| | - André S Mecawi
- Laboratory of Neuroendocrinology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 862, Edifício de Ciências Biomédicas, 7° andar, Vila Clementino, São Paulo, CEP 04023-062, Brasil.
| |
Collapse
|
9
|
Translational Studies in the Complex Role of Neurotransmitter Systems in Anxiety and Anxiety Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1191:121-140. [PMID: 32002926 DOI: 10.1007/978-981-32-9705-0_8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Discovery of innovative anxiolytics is severely hampering. Existing anxiolytics are developed decades ago and are still the therapeutics of choice. Moreover, lack of new drug targets forecasts a severe jeopardy in the future treatment of the huge population of CNS-diseased patients. We simply lack the knowledge on what is wrong in brains of anxious people (normal and diseased). Translational research, based on interacting clinical and preclinical research, is extremely urgent. In this endeavor, genetic and genomic approaches are part of the spectrum of contributing factors. We focus on three druggable targets: serotonin transporter, 5-HT1A, and GABAA receptors. It is still uncertain whether and how these targets are involved in normal and diseased anxiety processes. For serotonergic anxiolytics, the slow onset of action points to indirect effects leading to plasticity changes in brain systems leading to reduced anxiety. For GABAA benzodiazepine drugs, acute anxiolytic effects are found indicating primary mechanisms directly influencing anxiety processes. Close translational collaboration between fundamental academic and discovery research will lead to badly needed breakthroughs in the search for new anxiolytics.
Collapse
|
10
|
Dopamine D 2L Receptor Deficiency Causes Stress Vulnerability through 5-HT 1A Receptor Dysfunction in Serotonergic Neurons. J Neurosci 2019; 39:7551-7563. [PMID: 31371425 DOI: 10.1523/jneurosci.0079-19.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/16/2019] [Accepted: 05/28/2019] [Indexed: 12/27/2022] Open
Abstract
Mental disorders are caused by genetic and environmental factors. We here show that deficiency of an isoform of dopamine D2 receptor (D2R), D2LR, causes stress vulnerability in mouse. This occurs through dysfunction of serotonin [5-hydroxytryptamine (5-HT)] 1A receptor (5-HT1AR) on serotonergic neurons in the mouse brain. Exposure to forced swim stress significantly increased anxiety- and depressive-like behaviors in D2LR knock-out (KO) male mice compared with wild-type mice. Treatment with 8-OH-DPAT, a 5-HT1AR agonist, failed to alleviate the stress-induced behaviors in D2LR-KO mice. In forced swim-stressed D2LR-KO mice, 5-HT efflux in the medial prefrontal cortex was elevated and the expression of genes related to 5-HT levels was upregulated by the transcription factor PET1 in the dorsal raphe nucleus. Notably, D2LR formed a heteromer with 5-HT1AR in serotonergic neurons, thereby suppressing 5-HT1AR-activated G-protein-activated inwardly rectifying potassium conductance in D2LR-KO serotonergic neurons. Finally, D2LR overexpression in serotonergic neurons in the dorsal raphe nucleus alleviated stress vulnerability observed in D2LR-KO mice. Together, we conclude that disruption of the negative feedback regulation by the D2LR/5-HT1A heteromer causes stress vulnerability.SIGNIFICANCE STATEMENT Etiologies of mental disorders are multifactorial, e.g., interactions between genetic and environmental factors. In this study, using a mouse model, we showed that genetic depletion of an isoform of dopamine D2 receptor, D2LR, causes stress vulnerability associated with dysfunction of serotonin 1A receptor, 5-HT1AR in serotonergic neurons. The D2LR/5-HT1AR inhibitory G-protein-coupled heteromer may function as a negative feedback regulator to suppress psychosocial stress.
Collapse
|
11
|
Russo AM, Lawther AJ, Prior BM, Isbel L, Somers WG, Lesku JA, Richdale AL, Dissanayake C, Kent S, Lowry CA, Hale MW. Social approach, anxiety, and altered tryptophan hydroxylase 2 activity in juvenile BALB/c and C57BL/6J mice. Behav Brain Res 2018; 359:918-926. [PMID: 29935278 DOI: 10.1016/j.bbr.2018.06.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/24/2018] [Accepted: 06/19/2018] [Indexed: 12/28/2022]
Abstract
Autism spectrum disorder (ASD) is a heterogeneous and highly heritable condition with multiple aetiologies. Although the biological mechanisms underlying ASD are not fully understood, evidence suggests that dysregulation of serotonergic systems play an important role in ASD psychopathology. Preclinical models using mice with altered serotonergic neurotransmission may provide insight into the role of serotonin in behaviours relevant to clinical features of ASD. For example, BALB/c mice carry a loss-of-function single nucleotide polymorphism (SNP; C1473 G) in tryptophan hydroxylase 2 (Tph2), which encodes the brain-specific isoform of the rate-limiting enzyme for serotonin synthesis, and these mice frequently have been used to model symptoms of ASD. In this study, juvenile male BALB/c (G/G; loss-of-function variant) and C57BL/6 J (C/C; wild type variant) mice, were exposed to the three-chamber sociability test, and one week later to the elevated plus-maze (EPM). Tryptophan hydroxylase 2 (TPH2) activity was measured following injection of the aromatic amino acid decarboxylase (AADC)-inhibitor, NSD-1015, and subsequent HPLC detection of 5-hydroxytryptophan (5-HTP) within subregions of the dorsal raphe nucleus (DR) and median raphe nucleus (MnR). The BALB/c mice showed reduced social behaviour and increased anxious behaviour, as well as decreased 5-HTP accumulation in the rostral and mid-rostrocaudal DR. In the full cohort of mice, TPH2 activity in the mid-rostrocaudal DR was correlated with anxious behaviour in the EPM, however these correlations were not statistically significant within each strain, suggesting that TPH2 activity was not directly associated with either anxiety or sociability. Further research is therefore required to more fully understand how serotonergic systems are involved in mouse behaviours that resemble some of the clinical features of ASD.
Collapse
Affiliation(s)
- Adrian M Russo
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Adam J Lawther
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Benjamin M Prior
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Luke Isbel
- School of Molecular Sciences, La Trobe University, Melbourne, VIC, 3086, Australia
| | - W Gregory Somers
- Olga Tennison Autism Research Centre, La Trobe University, Melbourne, VIC, 3086, Australia
| | - John A Lesku
- School of Life Sciences, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Amanda L Richdale
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia; Olga Tennison Autism Research Centre, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Cheryl Dissanayake
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia; Olga Tennison Autism Research Centre, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Stephen Kent
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Christopher A Lowry
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Matthew W Hale
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
12
|
Huang JH, Chang HA, Fang WH, Ho PS, Liu YP, Wan FJ, Tzeng NS, Shyu JF, Chang CC. Serotonin receptor 1A promoter polymorphism, rs6295, modulates human anxiety levels via altering parasympathetic nervous activity. Acta Psychiatr Scand 2018; 137:263-272. [PMID: 29363117 DOI: 10.1111/acps.12853] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/02/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The G-allele of the -1019C/G (rs6295) promoter polymorphism of the serotonin receptor 1A (HTR1A) gene has been implicated in anxiety; however, the underlying neurophysiological processes are still not fully understood. Recent evidence indicates that low parasympathetic (vagal) tone is predictive of anxiety. We thus conducted a structural equation model (SEM) to examine whether the HTR1A rs6295 variant can affect anxiety by altering parasympathetic nervous activity. METHOD A sample of 1141 drug-free healthy Han Chinese was recruited for HTR1A genotyping. Autonomic nervous function was assessed by short-term spectral analysis of heart rate variability (HRV). Anxiety and stress levels were evaluated by the Beck Anxiety Inventory (BAI) and the Perceived Stress Scale (PSS) respectively. RESULTS The number of the HTR1A G allele was inversely correlated with high-frequency power (HF), a parasympathetic index of HRV. The HF index was negatively associated with BAI scores. Furthermore, the good-fitting SEM, adjusting for confounding variables (e.g., age and PSS levels), revealed a significant pathway linking rs6295 variant to BAI scores via HF index modulation. CONCLUSION These results are the first to show that HTR1A -1019C/G polymorphism influences anxiety levels by modulating parasympathetic tone, providing a neurophysiological insight into the role of HTR1A in human anxiety.
Collapse
Affiliation(s)
- J-H Huang
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - H-A Chang
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - W-H Fang
- Department of Family and Community Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - P-S Ho
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Y-P Liu
- Laboratory of Cognitive Neuroscience, Departments of Physiology and Psychiatry, National Defense Medical Center, Taipei, Taiwan
| | - F-J Wan
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - N-S Tzeng
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - J-F Shyu
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - C-C Chang
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
13
|
Pilar-Cuéllar F, Vidal R, Díaz Á, Garro-Martínez E, Linge R, Castro E, Haberzettl R, Fink H, Bert B, Brosda J, Romero B, Crespo-Facorro B, Pazos Á. Enhanced Stress Response in 5-HT 1AR Overexpressing Mice: Altered HPA Function and Hippocampal Long-Term Potentiation. ACS Chem Neurosci 2017; 8:2393-2401. [PMID: 28777913 DOI: 10.1021/acschemneuro.7b00156] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Postsynaptic 5-HT1A receptors (5-HT1AR) play an important role in anxiety and stress, although their contribution is still controversial. Previous studies report that mice overexpressing postsynaptic 5-HT1ARs show no changes in basal anxiety, though the influence of stress conditions has not been addressed yet. In this study, we used this animal model to evaluate the role of 5-HT1ARs in anxiety response after pre-exposure to an acute stressor. Under basal conditions, 5-HT1AR overexpressing animals presented high corticosterone levels and a lower mineralocorticoid/glucocorticoid receptor ratio. After pre-exposure to a single stressor, they showed a high anxiety-like response, associated with a blunted increase in corticosterone levels and higher c-Fos activation in the prefrontal cortex. Moreover, these mice also presented a lack of downregulation of hippocampal long-term potentiation after stress exposure. Therefore, higher postsynaptic 5-HT1AR activation might predispose to a high anxious phenotype and an impaired stress coping behavior.
Collapse
Affiliation(s)
- Fuencisla Pilar-Cuéllar
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Instituto
de Biomedicina y Biotecnologı́a de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, 39011 Santander, Spain
- Departamento
de Fisiología y Farmacología, Universidad de Cantabria, 39011 Santander, Spain
| | - Rebeca Vidal
- Departamento
de Farmacología, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre, 28041 Madrid, Spain
- Red de Trastornos Adictivos del Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Álvaro Díaz
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Instituto
de Biomedicina y Biotecnologı́a de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, 39011 Santander, Spain
- Departamento
de Fisiología y Farmacología, Universidad de Cantabria, 39011 Santander, Spain
| | - Emilio Garro-Martínez
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Instituto
de Biomedicina y Biotecnologı́a de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, 39011 Santander, Spain
- Departamento
de Fisiología y Farmacología, Universidad de Cantabria, 39011 Santander, Spain
| | - Raquel Linge
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Instituto
de Biomedicina y Biotecnologı́a de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, 39011 Santander, Spain
- Departamento
de Fisiología y Farmacología, Universidad de Cantabria, 39011 Santander, Spain
| | - Elena Castro
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Instituto
de Biomedicina y Biotecnologı́a de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, 39011 Santander, Spain
- Departamento
de Fisiología y Farmacología, Universidad de Cantabria, 39011 Santander, Spain
| | - Robert Haberzettl
- Institut
für Pharmakologie und Toxikologie, Fachbereich Veterinärmedizin, Freie Universität Berlin, 14195 Berlin, Germany
| | - Heidrun Fink
- Institut
für Pharmakologie und Toxikologie, Fachbereich Veterinärmedizin, Freie Universität Berlin, 14195 Berlin, Germany
| | - Bettina Bert
- Institut
für Pharmakologie und Toxikologie, Fachbereich Veterinärmedizin, Freie Universität Berlin, 14195 Berlin, Germany
| | - Jan Brosda
- Institut
für Pharmakologie und Toxikologie, Fachbereich Veterinärmedizin, Freie Universität Berlin, 14195 Berlin, Germany
| | - Beatriz Romero
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Instituto
de Biomedicina y Biotecnologı́a de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, 39011 Santander, Spain
- Departamento
de Fisiología y Farmacología, Universidad de Cantabria, 39011 Santander, Spain
| | - Benedicto Crespo-Facorro
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Hospital Universitario Marqués de Valdecilla, University of Cantabria-IDIVAL, School of Medicine, Department of Psychiatry, 39008 Santander, Spain
| | - Ángel Pazos
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Santander, Spain
- Instituto
de Biomedicina y Biotecnologı́a de Cantabria (IBBTEC), Universidad de Cantabria-CSIC-SODERCAN, 39011 Santander, Spain
- Departamento
de Fisiología y Farmacología, Universidad de Cantabria, 39011 Santander, Spain
| |
Collapse
|
14
|
Barrera-Bugueño C, Realini O, Escobar-Luna J, Sotomayor-Zárate R, Gotteland M, Julio-Pieper M, Bravo JA. Anxiogenic effects of a Lactobacillus, inulin and the synbiotic on healthy juvenile rats. Neuroscience 2017; 359:18-29. [DOI: 10.1016/j.neuroscience.2017.06.064] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 12/26/2022]
|
15
|
Wu ZM, Yang LH, Cui R, Ni GL, Wu FT, Liang Y. Contribution of Hippocampal 5-HT 3 Receptors in Hippocampal Autophagy and Extinction of Conditioned Fear Responses after a Single Prolonged Stress Exposure in Rats. Cell Mol Neurobiol 2017; 37:595-606. [PMID: 27324798 DOI: 10.1007/s10571-016-0395-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/13/2016] [Indexed: 02/01/2023]
Abstract
One of the hypotheses about the pathogenesis of posttraumatic stress disorder (PTSD) is the dysfunction of serotonin (5-HT) neurotransmission. While certain 5-HT receptor subtypes are likely critical for the symptoms of PTSD, few studies have examined the role of 5-HT3 receptor in the development of PTSD, even though 5-HT3 receptor is critical for contextual fear extinction and anxiety-like behavior. Therefore, we hypothesized that stimulation of 5-HT3 receptor in the dorsal hippocampus (DH) could prevent hippocampal autophagy and the development of PTSD-like behavior in animals. To this end, we infused SR57227, selective 5-HT3 agonist, into the DH after a single prolonged stress (SPS) treatment in rats. Three weeks later, we evaluated the effects of this pharmacological treatment on anxiety-related behaviors and extinction of contextual fear memory. We also accessed hippocampal autophagy and the expression of 5-HT3A subunit, Beclin-1, LC3-I, and LC3-II in the DH. We found that SPS treatment did not alter anxiety-related behaviors but prolonged the extinction of contextual fear memory, and such a behavioral phenomenon was correlated with increased hippocampal autophagy, decreased 5-HT3A expression, and increased expression of Beclin-1 and LC3-II/LC3-I ratio in the DH. Furthermore, intraDH infusions of SR57227 dose-dependently promoted the extinction of contextual fear memory, prevented hippocampal autophagy, and decreased expression of Beclin-1 and LC3-II/LC3-I ratio in the DH. These results indicated that 5-HT3 receptor in the hippocampus may play a critical role in the pathogenesis of hippocampal autophagy, and is likely involved in the pathophysiology of PTSD.
Collapse
Affiliation(s)
- Zhong-Min Wu
- Department of Anatomy, Medical College of Taizhou University, 1139 Taizhou city government Avenue, Taizhou, 318000, China
- Department of Neurology, First People's Hospital of Linhai City, Linhai, 317000, China
| | - Li-Hua Yang
- Department of Neurology, Taizhou Hospital, Taizhou, 317000, China
| | - Rong Cui
- Department of Neurology, First People's Hospital of Linhai City, Linhai, 317000, China
| | - Gui-Lian Ni
- Department of Neurology, First People's Hospital of Linhai City, Linhai, 317000, China
| | - Feng-Tian Wu
- City College of Zhejiang University, Hangzhou, 310015, China
| | - Yong Liang
- Department of Anatomy, Medical College of Taizhou University, 1139 Taizhou city government Avenue, Taizhou, 318000, China.
| |
Collapse
|
16
|
Noto B, Klempin F, Alenina N, Bader M, Fink H, Sander SE. Increased adult neurogenesis in mice with a permanent overexpression of the postsynaptic 5-HT 1A receptor. Neurosci Lett 2016; 633:246-251. [DOI: 10.1016/j.neulet.2016.09.051] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/25/2016] [Accepted: 09/09/2016] [Indexed: 02/07/2023]
|
17
|
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
|
18
|
Lakehayli S, Said N, El Khachibi M, El Ouahli M, Nadifi S, Hakkou F, Tazi A. Prenatal stress alters diazepam withdrawal syndrome and 5HT1A receptor expression in the raphe nuclei of adult rats. Neuroscience 2016; 330:50-6. [DOI: 10.1016/j.neuroscience.2016.05.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 05/17/2016] [Accepted: 05/17/2016] [Indexed: 10/21/2022]
|
19
|
Concordance and incongruence in preclinical anxiety models: Systematic review and meta-analyses. Neurosci Biobehav Rev 2016; 68:504-529. [PMID: 27328783 DOI: 10.1016/j.neubiorev.2016.04.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/19/2016] [Accepted: 04/18/2016] [Indexed: 12/14/2022]
Abstract
Rodent defense behavior assays have been widely used as preclinical models of anxiety to study possibly therapeutic anxiety-reducing interventions. However, some proposed anxiety-modulating factors - genes, drugs and stressors - have had discordant effects across different studies. To reconcile the effect sizes of purported anxiety factors, we conducted systematic review and meta-analyses of the literature on ten anxiety-linked interventions, as examined in the elevated plus maze, open field and light-dark box assays. Diazepam, 5-HT1A receptor gene knockout and overexpression, SERT gene knockout and overexpression, pain, restraint, social isolation, corticotropin-releasing hormone and Crhr1 were selected for review. Eight interventions had statistically significant effects on rodent anxiety, while Htr1a overexpression and Crh knockout did not. Evidence for publication bias was found in the diazepam, Htt knockout, and social isolation literatures. The Htr1a and Crhr1 results indicate a disconnect between preclinical science and clinical research. Furthermore, the meta-analytic data confirmed that genetic SERT anxiety effects were paradoxical in the context of the clinical use of SERT inhibitors to reduce anxiety.
Collapse
|
20
|
Sahli ZT, Banerjee P, Tarazi FI. The Preclinical and Clinical Effects of Vilazodone for the Treatment of Major Depressive Disorder. Expert Opin Drug Discov 2016; 11:515-23. [PMID: 26971593 PMCID: PMC4841022 DOI: 10.1517/17460441.2016.1160051] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Major depressive disorder (MDD) is the leading cause of disability worldwide, and according to the STAR*D trial, only 33% of patients with MDD responded to initial drug therapy. Augmentation of the leading class of antidepressant treatment, selective serotonin reuptake inhibitors (SSRIs), with the 5-HT1A receptor agonist buspirone has been shown to be effective in treating patients that do not respond to initial SSRI therapy. This suggests that newer treatments may improve the clinical picture of MDD. The US Food and Drug Administration (FDA) approved the antidepressant drug vilazodone (EMD 68843), a novel SSRI and 5-HT1A receptor partial agonist. Vilazodone has a half-life between 20-24 hours, reaches peak plasma concentrations at 3.7-5.3 hours, and is primarily metabolized by the hepatic CYP450 3A4 enzyme system. AREAS COVERED The authors review the preclinical and clinical profile of vilazodone. The roles of serotonin, the 5-HT1A receptor, and current pharmacotherapy approaches for MDD are briefly reviewed. Next, the preclinical pharmacological, behavioral, and physiological effects of vilazodone are presented, followed by the pharmacokinetic properties and metabolism of vilazodone in humans. Last, a brief summary of the main efficacy, safety, and tolerability outcomes of clinical trials of vilazodone is provided. EXPERT OPINION Vilazodone has shown efficacy versus placebo in improving depression symptoms in several double-blind, placebo-controlled trials. The long-term safety and tolerability of vilazodone treatment has also been established. Further studies are needed that directly compare patients treated with an SSRI (both with and without an adjunctive 5-HT1A partial agonist) versus patients treated with vilaozodone.
Collapse
Affiliation(s)
- Zeyad T Sahli
- a Department of Psychiatry and Neuroscience Program , Harvard Medical School, McLean Hospital , Belmont , MA , USA.,b School of Medicine , American University of Beirut , Beirut , Lebanon
| | - Pradeep Banerjee
- c Forest Research Institute , Jersey City , NJ , USA , an affiliate of Allergan Inc
| | - Frank I Tarazi
- a Department of Psychiatry and Neuroscience Program , Harvard Medical School, McLean Hospital , Belmont , MA , USA
| |
Collapse
|
21
|
Twarkowski H, Hagena H, Manahan-Vaughan D. The 5-hydroxytryptamine4 receptor enables differentiation of informational content and encoding in the hippocampus. Hippocampus 2016; 26:875-91. [PMID: 26800645 PMCID: PMC5067691 DOI: 10.1002/hipo.22569] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 01/15/2016] [Accepted: 01/20/2016] [Indexed: 11/10/2022]
Abstract
Long‐term synaptic plasticity, represented by long‐term depression (LTD) and long‐term potentiation (LTP) comprise cellular processes that enable memory. Neuromodulators such as serotonin regulate hippocampal function, and the 5‐HT4‐receptor contributes to processes underlying cognition. It was previously shown that in the CA1‐region, 5‐HT4‐receptors regulate the frequency‐response relationship of synaptic plasticity: patterned afferent stimulation that has no effect on synaptic strength (i.e., a θm‐frequency), will result in LTP or LTD, when given in the presence of a 5‐HT4‐agonist, or antagonist, respectively. Here, we show that in the dentate gyrus (DG) and CA3 regions of freely behaving rats, pharmacological manipulations of 5‐HT4‐receptors do not influence responses generated at θm‐frequencies, but activation of 5‐HT4‐receptors prevents persistent LTD in mossy fiber (mf)‐CA3, or perforant path‐DG synapses. Furthermore, the regulation by 5‐HT4‐receptors of LTP is subfield‐specific: 5‐HT4‐receptor‐activation prevents mf‐CA3‐LTP, but does not strongly affect DG‐potentiation. These data suggest that 5‐HT4‐receptor activation prioritises information encoding by means of LTP in the DG and CA1 regions, and suppresses persistent information storage in mf‐CA3 synapses. Thus, 5‐HT4‐receptors serve to shape information storage across the hippocampal circuitry and specify the nature of experience‐dependent encoding. © 2016 The Authors Hippocampus Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hannah Twarkowski
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Hardy Hagena
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Denise Manahan-Vaughan
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
22
|
Kiryanova V, Meunier SJ, Vecchiarelli HA, Hill MN, Dyck RH. Effects of maternal stress and perinatal fluoxetine exposure on behavioral outcomes of adult male offspring. Neuroscience 2016; 320:281-96. [PMID: 26872999 DOI: 10.1016/j.neuroscience.2016.01.064] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/20/2016] [Accepted: 01/29/2016] [Indexed: 12/25/2022]
Abstract
UNLABELLED Women of child-bearing age are the population group at highest risk for depression. In pregnant women, fluoxetine (Flx) is the most widely prescribed selective serotonin reuptake inhibitor (SSRI) used for the treatment of depression. While maternal stress, depression, and Flx exposure have been shown to effect neurodevelopment of the offspring, separately, combined effects of maternal stress and Flx exposure have not been extensively examined. The present study investigated the effects of prenatal maternal stress and perinatal exposure to the SSRI Flx on the behavior of male mice as adults. METHODS C57BL/6 dams exposed to chronic unpredictable stress from embryonic (E) day 4 to E18 and non-stressed dams were administered Flx (25 mg/kg/d) in the drinking water from E15 to postnatal day 12. A separate control group consisted of animals that were not exposed to stress or Flx. At 12 days of age, brain levels of serotonin were assessed in the male offspring. At two months of age, the male offspring of mothers exposed to prenatal stress (PS), perinatal Flx, PS and Flx, or neither PS or Flx, went through a comprehensive behavioral test battery. At the end of testing brain-derived neurotropic factor (BDNF) levels were assessed in the frontal cortex of the offspring. RESULTS Maternal behavior was not altered by either stress or Flx treatment. Treatment of the mother with Flx led to detectible Flx and NorFlx levels and lead to a decrease in serotonin levels in pup brains. In the adult male offspring, while perinatal exposure to Flx increased aggressive behavior, prenatal maternal stress decreased aggressive behavior. Interestingly, the combined effects of stress and Flx normalized aggressive behavior. Furthermore, perinatal Flx treatment led to a decrease in anxiety-like behavior in male offspring. PS led to hyperactivity and a decrease in BDNF levels in the frontal cortex regardless of Flx exposure. Neither maternal stress or Flx altered offspring performance in tests of cognitive abilities, memory, sensorimotor information processing, or risk assessment behaviors. These results demonstrate that maternal exposure to stress and Flx have a number of sustained effects on the male offspring.
Collapse
Affiliation(s)
- V Kiryanova
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - S J Meunier
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - H A Vecchiarelli
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - M N Hill
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - R H Dyck
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
23
|
Wu ZM, Zheng CH, Zhu ZH, Wu FT, Ni GL, Liang Y. SiRNA-mediated serotonin transporter knockdown in the dorsal raphe nucleus rescues single prolonged stress-induced hippocampal autophagy in rats. J Neurol Sci 2015; 360:133-40. [PMID: 26723990 DOI: 10.1016/j.jns.2015.11.056] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 11/26/2015] [Accepted: 11/30/2015] [Indexed: 02/02/2023]
Abstract
The neurobiological mechanisms underlying the development of post-traumatic stress disorder (PTSD) remain elusive. One of the hypotheses is the dysfunction of serotonin (5-HT) neurotransmission, which is critically regulated by serotonin transporter (SERT). Therefore, we hypothesized that attenuation of SERT gene expression in the hippocampus could prevent hippocampal autophagy and the development of PTSD-like behavior. To this end, we infused SLC6A4 siRNAs into the dorsal raphe nucleus (DRN) to knockdown SERT gene expression after a single prolonged stress (SPS) treatment in rats. Then, we evaluated the effects of SERT gene knockdown on anxiety-related behaviors and extinction of contextual fear memory. We also examined the histological changes and the expression of Beclin-1, LC3-I, and LC3-II in the hippocampus. We found that SPS treatment did not alter anxiety-related behaviors but prolonged the extinction of contextual fear memory, and such a behavioral phenomenon was correlated with increased hippocampal autophagy, decreased 5-HT level, and increased expression of Beclin-1 and LC3-II/LC3-I ratio in the hippocampus. Furthermore, intra-DRN infusion of SLC6A4 siRNAs promoted the extinction of contextual fear memory, prevented hippocampal autophagy, increased 5-HT level, and decreased expression of Beclin-1 and LC3-II/LC3-I ratio. These results indicated that SERT may play a critical role in the pathogenesis of hippocampal autophagy, and is likely involved in the development of PTSD.
Collapse
Affiliation(s)
- Zhong-Min Wu
- Department of anatomy, Medical College of Taizhou University, Taizhou 318000, China; Department of Neurology, First People's Hospital of Linhai City, Linhai 317000, China
| | - Chun-Hua Zheng
- Outpatient Office, Taizhou Hospital, Taizhou 317000, China
| | - Zhen-Hua Zhu
- Department of Pediatrics, Taizhou Central Hospital, Taizhou 318000, China
| | - Feng-Tian Wu
- City Colloege of Zhejiang University, Hanzhou 310031, China
| | - Gui-Lian Ni
- Department of Neurology, First People's Hospital of Linhai City, Linhai 317000, China
| | - Yong Liang
- Department of anatomy, Medical College of Taizhou University, Taizhou 318000, China.
| |
Collapse
|
24
|
Ohno Y, Shimizu S, Tokudome K, Kunisawa N, Sasa M. New insight into the therapeutic role of the serotonergic system in Parkinson's disease. Prog Neurobiol 2015; 134:104-21. [DOI: 10.1016/j.pneurobio.2015.09.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 08/17/2015] [Accepted: 09/05/2015] [Indexed: 11/30/2022]
|
25
|
Chilmonczyk Z, Bojarski AJ, Pilc A, Sylte I. Functional Selectivity and Antidepressant Activity of Serotonin 1A Receptor Ligands. Int J Mol Sci 2015; 16:18474-506. [PMID: 26262615 PMCID: PMC4581256 DOI: 10.3390/ijms160818474] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 07/29/2015] [Accepted: 07/31/2015] [Indexed: 01/11/2023] Open
Abstract
Serotonin (5-HT) is a monoamine neurotransmitter that plays an important role in physiological functions. 5-HT has been implicated in sleep, feeding, sexual behavior, temperature regulation, pain, and cognition as well as in pathological states including disorders connected to mood, anxiety, psychosis and pain. 5-HT1A receptors have for a long time been considered as an interesting target for the action of antidepressant drugs. It was postulated that postsynaptic 5-HT1A agonists could form a new class of antidepressant drugs, and mixed 5-HT1A receptor ligands/serotonin transporter (SERT) inhibitors seem to possess an interesting pharmacological profile. It should, however, be noted that 5-HT1A receptors can activate several different biochemical pathways and signal through both G protein-dependent and G protein-independent pathways. The variables that affect the multiplicity of 5-HT1A receptor signaling pathways would thus result from the summation of effects specific to the host cell milieu. Moreover, receptor trafficking appears different at pre- and postsynaptic sites. It should also be noted that the 5-HT1A receptor cooperates with other signal transduction systems (like the 5-HT1B or 5-HT2A/2B/2C receptors, the GABAergic and the glutaminergic systems), which also contribute to its antidepressant and/or anxiolytic activity. Thus identifying brain specific molecular targets for 5-HT1A receptor ligands may result in a better targeting, raising a hope for more effective medicines for various pathologies.
Collapse
Affiliation(s)
- Zdzisław Chilmonczyk
- National Medicines Institute, Chełmska 30/34, 00-725 Warszawa, Poland.
- Institute of Nursing and Health Sciences, University of Rzeszów, W. Kopisto 2A, 35-310 Rzeszów, Poland.
| | - Andrzej Jacek Bojarski
- Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Kraków, Poland.
| | - Andrzej Pilc
- Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Kraków, Poland.
| | - Ingebrigt Sylte
- Faculty of Health Sciences, University of Tromsø-The Arctic University of Norway, No-9037 Tromsø, Norway.
| |
Collapse
|
26
|
Environmental Enrichment Reduces Anxiety by Differentially Activating Serotonergic and Neuropeptide Y (NPY)-Ergic System in Indian Field Mouse (Mus booduga): An Animal Model of Post-Traumatic Stress Disorder. PLoS One 2015; 10:e0127945. [PMID: 26016844 PMCID: PMC4446351 DOI: 10.1371/journal.pone.0127945] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 04/20/2015] [Indexed: 11/19/2022] Open
Abstract
Exposure to a predator elicits an innate fear response and mimics several behavioral disorders related to post-traumatic stress disorder (PTSD). The protective role of an enriched condition (EC) against psychogenic stressors in various animal models has been well documented. However, this condition has not been tested in field mice in the context of PTSD. In this study, we show that field mice (Mus booduga) housed under EC exhibit predominantly proactive and less reactive behavior compared with mice housed under standard conditions (SC) during exposure to their natural predator (field rat Rattus rattus). Furthermore, we observed that EC mice displayed less anxiety-like behavior in an elevated plus maze (EPM) and light/dark-box after exposure to the predator (7 hrs/7 days). In EC mice, predator exposure elevated the level of serotonin (5-Hydroxytrypamine, [5-HT]) in the amygdala as part of the coping response. Subsequently, the serotonin transporter (SERT) and 5-HT1A receptor were up-regulated significantly, but the same did not occur in the 5-HT2C receptor, which is associated with the activation of calmodulin-dependent protein kinase-II (CaMKII) and a transcription factor cAMP response element binding protein (CREB). Our results show that predator exposure induced the activation of CaMKII/CREB, which is accompanied with increased levels of histone acetylation (H3, H4) and decreased histone deacetylases (HDAC1, 2). Subsequently, in the amygdala, the transcription of brain-derived neurotrophic factor (BDNF), neuropeptide Y (NPY) and its Y1 receptor were up-regulated, whereas the Y2 receptor was down-regulated. Therefore, EC facilitated a coping response against a fear associated cue in a PTSD animal model and reduced anxiety by differentially activating serotonergic and NPY-ergic systems.
Collapse
|
27
|
Zohar I, Dosoretz-Abittan L, Shoham S, Weinstock M. Sex dependent reduction by prenatal stress of the expression of 5HT1A receptors in the prefrontal cortex and CRF type 2 receptors in the raphe nucleus in rats: reversal by citalopram. Psychopharmacology (Berl) 2015; 232:1643-53. [PMID: 25420605 DOI: 10.1007/s00213-014-3803-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/27/2014] [Indexed: 01/10/2023]
Abstract
RATIONALE Alterations in the serotonergic transmission and activity of corticotropin-releasing factor (CRF) family may underlie anxiety and depressive disorders. These could be corrected by treatment with SSRIs. OBJECTIVES The objective of the current study is to determine whether the increased anxiety of prenatally stressed (PS) rats of both sexes is associated with changes in 5HT1A and CRF type 2 receptors (5HT1AR and CRFR2) in the prefrontal cortex (PFC)-dorsal raphe nuclei (DRN) axis, and how these are affected by chronic treatment with citalopram (10 mg/kg/day). We focussed on GABAergic cells that co-express parvalbumin and/or neuropeptide Y, and 5HT1AR in the medial prefrontal cortex (mPFC) and on cells that express 5HT, parvalbumin, 5HT1AR or CRFR2 in the DRN. RESULTS Immunohistochemistry with fluorescent antibodies demonstrated sex differences in the expression of 5HT1AR and CRFR2 protein. Prenatal stress selectively reduced the expression of 5HT1AR on GABAergic cells in the mPFC in males and that of CRFR2 in the DRN of females. Citalopram treatment for 5 weeks abolished the increase in anxiety in both sexes, restored the intensity of expression of 5HT1AR in the mPFC in males and increased their expression in the mPFC and DRN in females. Citalopram reduced CRFR2 expression in control and PS males but increased it in PS females. CONCLUSIONS Male and female rats show differences in the expression of 5HT1AR and CRFR2 protein that are selectively reduced by prenatal stress. Reversal by citalopram of the changes in the expression of these receptors induced by prenatal stress support their role in the aetiology of anxiety.
Collapse
Affiliation(s)
- Inbar Zohar
- Department of Pharmacology, Institute of Drug Research, Hebrew University Medical Centre, Ein Kerem, Jerusalem, 91120, Israel
| | | | | | | |
Collapse
|
28
|
Albert PR, Fiori LM. Transcriptional dys-regulation in anxiety and major depression: 5-HT1A gene promoter architecture as a therapeutic opportunity. Curr Pharm Des 2015; 20:3738-50. [PMID: 24180393 DOI: 10.2174/13816128113196660740] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/23/2013] [Indexed: 12/31/2022]
Abstract
The etiology of major depression remains unclear, but reduced activity of the serotonin (5-HT) system remains implicated and treatments that increase 5-HT neurotransmission can ameliorate depressive symptoms. 5-HT1A receptors are critical regulators of the 5- HT system. They are expressed as both presynaptic autoreceptors that negatively regulate 5-HT neurons, and as post-synaptic heteroreceptors on non-serotonergic neurons in the hippocampus, cortex, and limbic system that are critical to mediate the antidepressant actions of 5-HT. Thus, 5-HT1A auto- and heteroreceptors have opposite actions on serotonergic neurotransmission. Because most 5-HT1A ligands target both auto- and heteroreceptors their efficacy has been limited, resulting in weak or unclear responses. We propose that by understanding the transcriptional regulation of the 5-HT1A receptor it may be possible to regulate its expression differentially in raphe and projection regions. Here we review the transcriptional architecture of the 5-HT1A gene (HTR1A) with a focus on specific DNA elements and transcription factors that have been shown to regulate 5-HT1A receptor expression in the brain. Association studies with the functional HTR1A promoter polymorphism rs6295 suggest a new model for the role of the 5-HT1A receptor in susceptibility to depression involving early deficits in cognitive, fear and stress reactivity as stressors that may ultimately lead to depression. We present evidence that by targeting specific transcription factors it may be possible to oppositely regulate 5-HT1A auto- and heteroreceptor expression, synergistically increasing serotonergic neurotransmission for the treatment of depression.
Collapse
Affiliation(s)
| | - Laura M Fiori
- Ottawa Hospital Research Institute, Neuroscience, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, Canada K1H-8M5.
| |
Collapse
|
29
|
Tu W, Cook A, Scholl JL, Mears M, Watt MJ, Renner KJ, Forster GL. Serotonin in the ventral hippocampus modulates anxiety-like behavior during amphetamine withdrawal. Neuroscience 2014; 281:35-43. [PMID: 25241066 DOI: 10.1016/j.neuroscience.2014.09.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 09/06/2014] [Accepted: 09/10/2014] [Indexed: 12/13/2022]
Abstract
Withdrawal from amphetamine is associated with increased anxiety and sensitivity to stressors which are thought to contribute to relapse. Rats undergoing amphetamine withdrawal fail to exhibit stress-induced increases in serotonin (5-HT) release in the ventral hippocampus and show heightened anxiety-like behaviors. Therefore, we tested the hypothesis that reducing 5-HT levels in the ventral hippocampus is a causal mechanism in increasing anxiety-like behaviors during amphetamine withdrawal. First, we tested whether reducing 5-HT levels in the ventral hippocampus directly increases anxiety behavior. Male rats were bilaterally infused with 5,7-dihydroxytryptamine (5,7-DHT) into the ventral hippocampus, which produced a 83% decrease in ventral hippocampus 5-HT content, and were tested on the elevated plus maze (EPM) for anxiety-like behavior. Reducing ventral hippocampus 5-HT levels decreased the time spent in the open arms of the maze, suggesting that diminished ventral hippocampus 5-HT levels increases anxiety-like behavior. Next, we tested whether increasing 5-HT levels in the ventral hippocampus reverses anxiety behavior exhibited by rats undergoing amphetamine withdrawal. Rats were treated daily with either amphetamine (2.5-mg/kg, i.p.) or saline for 2weeks, and at 2weeks withdrawal, were infused with the selective serotonin reuptake inhibitor paroxetine (0.5μM) bilaterally into the ventral hippocampus and tested for anxiety-like behavior on the EPM. Rats pre-treated with amphetamine exhibited increased anxiety-like behavior on the EPM. This effect was reversed by ventral hippocampus infusion of paroxetine. Our results suggest that 5-HT levels in the ventral hippocampus are critical for regulating anxiety behavior. Increasing 5-HT levels during withdrawal may be an effective strategy for reducing anxiety-induced drug relapse.
Collapse
Affiliation(s)
- W Tu
- Center for Brain and Behavior Research, Division of Basic Biomedical Sciences, Sanford School of Medicine at the University of South Dakota, 414 East Clark Street, Vermillion, SD, USA
| | - A Cook
- Center for Brain and Behavior Research, Division of Basic Biomedical Sciences, Sanford School of Medicine at the University of South Dakota, 414 East Clark Street, Vermillion, SD, USA
| | - J L Scholl
- Center for Brain and Behavior Research, Division of Basic Biomedical Sciences, Sanford School of Medicine at the University of South Dakota, 414 East Clark Street, Vermillion, SD, USA
| | - M Mears
- Center for Brain and Behavior Research, Division of Basic Biomedical Sciences, Sanford School of Medicine at the University of South Dakota, 414 East Clark Street, Vermillion, SD, USA
| | - M J Watt
- Center for Brain and Behavior Research, Division of Basic Biomedical Sciences, Sanford School of Medicine at the University of South Dakota, 414 East Clark Street, Vermillion, SD, USA
| | - K J Renner
- Center for Brain and Behavior Research, Biology Department, University of South Dakota, 414 East Clark Street, Vermillion, SD, USA
| | - G L Forster
- Center for Brain and Behavior Research, Division of Basic Biomedical Sciences, Sanford School of Medicine at the University of South Dakota, 414 East Clark Street, Vermillion, SD, USA.
| |
Collapse
|
30
|
Albert PR, Vahid-Ansari F, Luckhart C. Serotonin-prefrontal cortical circuitry in anxiety and depression phenotypes: pivotal role of pre- and post-synaptic 5-HT1A receptor expression. Front Behav Neurosci 2014; 8:199. [PMID: 24936175 PMCID: PMC4047678 DOI: 10.3389/fnbeh.2014.00199] [Citation(s) in RCA: 202] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/16/2014] [Indexed: 01/03/2023] Open
Abstract
Decreased serotonergic activity has been implicated in anxiety and major depression, and antidepressants directly or indirectly increase the long-term activity of the serotonin system. A key component of serotonin circuitry is the 5-HT1A autoreceptor, which functions as the major somatodendritic autoreceptor to negatively regulate the "gain" of the serotonin system. In addition, 5-HT1A heteroreceptors are abundantly expressed post-synaptically in the prefrontal cortex (PFC), amygdala, and hippocampus to mediate serotonin actions on fear, anxiety, stress, and cognition. Importantly, in the PFC 5-HT1A heteroreceptors are expressed on at least two antagonist neuronal populations: excitatory pyramidal neurons and inhibitory interneurons. Rodent models implicate the 5-HT1A receptor in anxiety- and depression-like phenotypes with distinct roles for pre- and post-synaptic 5-HT1A receptors. In this review, we present a model of serotonin-PFC circuitry that integrates evidence from mouse genetic models of anxiety and depression involving knockout, suppression, over-expression, or mutation of genes of the serotonin system including 5-HT1A receptors. The model postulates that behavioral phenotype shifts as serotonin activity increases from none (depressed/aggressive not anxious) to low (anxious/depressed) to high (anxious, not depressed). We identify a set of conserved transcription factors including Deaf1, Freud-1/CC2D1A, Freud-2/CC2D1B and glucocorticoid receptors that may confer deleterious regional changes in 5-HT1A receptors in depression, and how future treatments could target these mechanisms. Further studies to specifically test the roles and regulation of pyramidal vs. interneuronal populations of 5-HT receptors are needed better understand the role of serotonin in anxiety and depression and to devise more effective targeted therapeutic approaches.
Collapse
Affiliation(s)
- Paul R Albert
- Neuroscience, Ottawa Hospital Research Institute, University of Ottawa Ottawa, ON, Canada
| | - Faranak Vahid-Ansari
- Neuroscience, Ottawa Hospital Research Institute, University of Ottawa Ottawa, ON, Canada ; Department of Cellular and Molecular Medicine, University of Ottawa Ottawa ON, Canada
| | - Christine Luckhart
- Neuroscience, Ottawa Hospital Research Institute, University of Ottawa Ottawa, ON, Canada ; Department of Cellular and Molecular Medicine, University of Ottawa Ottawa ON, Canada
| |
Collapse
|
31
|
Abdala AP, Bissonnette JM, Newman-Tancredi A. Pinpointing brainstem mechanisms responsible for autonomic dysfunction in Rett syndrome: therapeutic perspectives for 5-HT1A agonists. Front Physiol 2014; 5:205. [PMID: 24910619 PMCID: PMC4038922 DOI: 10.3389/fphys.2014.00205] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 05/12/2014] [Indexed: 12/26/2022] Open
Abstract
Rett syndrome is a neurological disorder caused by loss of function of methyl-CpG-binding protein 2 (MeCP2). Reduced function of this ubiquitous transcriptional regulator has a devastating effect on the central nervous system. One of the most severe and life-threatening presentations of this syndrome is brainstem dysfunction, which results in autonomic disturbances such as breathing deficits, typified by episodes of breathing cessation intercalated with episodes of hyperventilation or irregular breathing. Defects in numerous neurotransmitter systems have been observed in Rett syndrome both in animal models and patients. Here we dedicate special attention to serotonin due to its role in promoting regular breathing, increasing vagal tone, regulating mood, alleviating Parkinsonian-like symptoms and potential for therapeutic translation. A promising new symptomatic strategy currently focuses on regulation of serotonergic function using highly selective serotonin type 1A (5-HT1A) “biased agonists.” We address this newly emerging therapy for respiratory brainstem dysfunction and challenges for translation with a holistic perspective of Rett syndrome, considering potential mood and motor effects.
Collapse
Affiliation(s)
- Ana P Abdala
- School of Physiology and Pharmacology, University of Bristol Bristol, UK
| | - John M Bissonnette
- Department of Obstetrics and Gynecology, Oregon Health and Science University Portland, OR, USA
| | | |
Collapse
|
32
|
Bader LR, Carboni JD, Burleson CA, Cooper MA. 5-HT1A receptor activation reduces fear-related behavior following social defeat in Syrian hamsters. Pharmacol Biochem Behav 2014; 122:182-90. [PMID: 24726709 DOI: 10.1016/j.pbb.2014.03.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 03/03/2014] [Accepted: 03/30/2014] [Indexed: 12/25/2022]
Abstract
Social defeat leads to selective avoidance of familiar opponents as well as general avoidance of novel, non-threatening intruders. Avoidance of familiar opponents represents a fear-related memory whereas generalized social avoidance indicates anxiety-like behavior. We have previously shown that serotonin signaling alters responses to social defeat in Syrian hamsters, although it is unclear whether serotonin modulates defeat-induced fear, anxiety, or both. In this study we focus on 5-HT1A receptors, in part, because their activation had been linked to the acquisition of conditioned fear. We hypothesized that pharmacological activation of 5-HT1A receptors prior to social defeat would reduce avoidance of familiar opponents and impair Arc expression in the basolateral amygdala (BLA), but not alter anxiety-like behavior. We administered 8-OH-DPAT, a 5-HT1A receptor agonist, prior to 3, 5-minute social defeats and 24h later exposed hamsters to a social interaction test to measure the conditioned defeat response immediately followed by either a Y-maze test or an open field test. In a separate experiment, we administered 8-OH-DPAT prior to 3, 5-minute social defeats and later removed the brains for Arc immunohistochemistry. Social defeat increased the number of Arc immunopositive cells in the central amygdala (CeA), prelimbic cortex (PL), and BLA, and 8-OH-DPAT treatment reduced Arc immunoreactivity in the PL. These results suggest that 5-HT1A receptor activation impairs the fear memory associated with social defeat, but does not alter defeat-induced anxiety. Overall, 5-HT1A receptor activation may impair Arc expression in select brain regions such as the PL and thereby disrupt the development of a fear memory essential for the conditioned defeat response.
Collapse
Affiliation(s)
- Lauren R Bader
- Department of Psychology, University of Tennessee, Knoxville, TN 37996, USA.
| | - Joseph D Carboni
- Department of Psychology, University of Tennessee, Knoxville, TN 37996, USA
| | - Cody A Burleson
- Department of Psychology, University of Tennessee, Knoxville, TN 37996, USA
| | - Matthew A Cooper
- Department of Psychology, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
33
|
Garcia-Garcia A, Tancredi AN, Leonardo ED. 5-HT(1A) [corrected] receptors in mood and anxiety: recent insights into autoreceptor versus heteroreceptor function. Psychopharmacology (Berl) 2014; 231:623-36. [PMID: 24337875 PMCID: PMC3927969 DOI: 10.1007/s00213-013-3389-x] [Citation(s) in RCA: 161] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 11/26/2013] [Indexed: 11/26/2022]
Abstract
RATIONALE Serotonin (5-HT) neurotransmission is intimately linked to anxiety and depression and a diverse body of evidence supports the involvement of the main inhibitory serotonergic receptor, the serotonin-1A (5-HT(1A)) subtype, in both disorders. OBJECTIVES In this review, we examine the function of 5-HT(1A) receptor subpopulations and re-interpret our understanding of their role in mental illness in light of new data, separating both spatial (autoreceptor versus heteroreceptor) and the temporal (developmental versus adult) roles of the endogenous 5-HT(1A) receptors, emphasizing their distinct actions in mediating anxiety and depression-like behaviors. RESULTS It is difficult to unambiguously distinguish the effects of different populations of the 5-HT(1A) receptors with traditional genetic animal models and pharmacological approaches. However, with the advent of novel genetic systems and subpopulation-selective pharmacological agents, direct evidence for the distinct roles of these populations in governing emotion-related behavior is emerging. CONCLUSIONS There is strong and growing evidence for a functional dissociation between auto- and heteroreceptor populations in mediating anxiety and depressive-like behaviors, respectively. Furthermore, while it is well established that 5-HT(1A) receptors act developmentally to establish normal anxiety-like behaviors, the developmental role of 5-HT(1A) heteroreceptors is less clear, and the specific mechanisms underlying the developmental role of each subpopulation are likely to be key elements determining mood control in adult subjects.
Collapse
Affiliation(s)
- Alvaro Garcia-Garcia
- Department of Psychiatry, Division of Integrative Neuroscience, Columbia University and the New York State Psychiatric Institute, 1051 Riverside Dr. Box 87, New York, NY 10032
- Correspondence should be addressed to either AGG at or EDL at , Telephone: (001) 212-543-5266, Fax: (001) 212-543-5129
| | | | - E. David Leonardo
- Department of Psychiatry, Division of Integrative Neuroscience, Columbia University and the New York State Psychiatric Institute, 1051 Riverside Dr. Box 87, New York, NY 10032
- Correspondence should be addressed to either AGG at or EDL at , Telephone: (001) 212-543-5266, Fax: (001) 212-543-5129
| |
Collapse
|
34
|
Kim ER, Min BH, Lee TH, Son M, Rhee PL. Effect of DA-9701 on colorectal distension-induced visceral hypersensitivity in a rat model. Gut Liver 2014; 8:388-93. [PMID: 25071903 PMCID: PMC4113047 DOI: 10.5009/gnl.2014.8.4.388] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 05/22/2013] [Accepted: 07/07/2013] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND/AIMS DA-9701 is a newly developed drug made from the vegetal extracts of Pharbitidis semen and Co-rydalis tuber. The aim of this study was to evaluate the effect of DA-9701 on colorectal distension (CRD)-induced visceral hypersensitivity in a rat model. METHODS Male Sprague-Dawley rats were subjected to neonatal colon irritation (CI) using CRD at 1 week after birth (CI group). At 6 weeks after birth, CRD was applied to these rats with a pressure of 20 to 90 mm Hg, and changes in the mean arterial pressure (MAP) were measured at baseline (i.e., without any drug administration) and after the administration of different doses of DA-9701. RESULTS In the absence of DA-9701, the MAP changes after CRD were significantly higher in the CI group than in the control group at all applied pressures. In the control group, MAP changes after CRD were not significantly affected by the administration of DA-9701. In the CI group, however, the administration of DA-9701 resulted in a significant decrease in MAP changes after CRD. The administration of DA-9701 at a dose of 1.0 mg/kg produced a more significant decrease in MAP changes than the 0.3 mg/kg dose. CONCLUSIONS The administration of DA-9701 resulted in a significant increase in pain threshold in rats with CRD-induced visceral hypersensitivity.
Collapse
Affiliation(s)
- Eun Ran Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Byung-Hoon Min
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae Ho Lee
- Dong-A ST, Co., Ltd., Research Center, Yongin, Korea
| | - Miwon Son
- Dong-A ST, Co., Ltd., Research Center, Yongin, Korea
| | - Poong-Lyul Rhee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
35
|
Serotonin 5-HT1A receptors as targets for agents to treat psychiatric disorders: rationale and current status of research. CNS Drugs 2013; 27:703-16. [PMID: 23757185 DOI: 10.1007/s40263-013-0071-0] [Citation(s) in RCA: 230] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Psychiatric disorders represent a large economic burden in modern societies. However, pharmacological treatments are still far from optimal. Drugs used in the treatment of major depressive disorder (MDD) and anxiety disorders (selective serotonin [5-HT] reuptake inhibitors [SSRIs] and serotonin-noradrenaline reuptake inhibitors [SNRIs]) are pharmacological refinements of first-generation tricyclic drugs, discovered by serendipity, and show low efficacy and slowness of onset. Moreover, antipsychotic drugs are partly effective in positive symptoms of schizophrenia, yet they poorly treat negative symptoms and cognitive deficits. The present article reviews the neurobiological basis of 5-HT1A receptor (5-HT1A-R) function and the role of pre- and postsynaptic 5-HT1A-Rs in the treatment of MDD, anxiety and psychotic disorders. The activation of postsynaptic 5-HT1A-Rs in corticolimbic areas appears beneficial for the therapeutic action of antidepressant drugs. However, presynaptic 5-HT1A-Rs play a detrimental role in MDD, since individuals with high density or function of presynaptic 5-HT1A-Rs are more susceptible to mood disorders and suicide, and respond poorly to antidepressant drugs. Moreover, the indirect activation of presynaptic 5-HT1A-Rs by SSRIs/SNRIs reduces 5-HT neuron activity and terminal 5-HT release, thus opposing the elevation of extracellular 5-HT produced by blockade of the serotonin transporter (SERT) in the forebrain. Chronic antidepressant treatment desensitizes presynaptic 5-HT1A-Rs, thus reducing the effectiveness of the 5-HT1A autoreceptor-mediated negative feedback. The prevention of this process by the non-selective partial agonist pindolol accelerates clinical antidepressant effects. Two new antidepressant drugs, vilazodone (marketed in the USA) and vortioxetine (in development) incorporate partial 5-HT1A-R agonist properties with SERT blockade. Several studies with transgenic mice have also established the respective role of pre- and postsynaptic 5-HT1A-Rs in MDD and anxiety. In agreement with pharmacological studies, presynaptic and postsynaptic 5-HT1A-R activation appears necessary for anxiolytic and antidepressant effects, respectively, yet, neurodevelopmental roles for 5-HT1A-Rs are also involved. Likewise, the use of small interference RNA has enabled the showing of robust antidepressant-like effects in mice after selective knock-down of 5-HT1A autoreceptors. Postsynaptic 5-HT1A-Rs in the prefrontal cortex (PFC) also appear important for the superior clinical effects of clozapine and other second-generation (atypical) antipsychotic drugs in the treatment of schizophrenia and related psychotic disorders. Despite showing a moderate in vitro affinity for 5-HT1A-Rs in binding assays, clozapine displays functional agonist properties at this receptor type in vivo. The stimulation of 5-HT1A-Rs in the PFC leads to the distal activation of the mesocortical pathway and to an increased dopamine release in PFC, an effect likely involved in the clinical actions of clozapine in negative symptoms and cognitive deficits in schizophrenia. The anxiolytic/antidepressant properties of 5-HT1A-R agonists in preclinical tests raised expectations enormously. However, these agents have achieved little clinical success, possibly due to their partial agonist character at postsynaptic 5-HT1A-Rs, together with full agonist properties at presynaptic 5-HT1A autoreceptors, as well as their gastrointestinal side effects. The partial 5-HT1A-R agonists buspirone, gepirone, and tandospirone are marketed as anxiolytic drugs, and buspirone is also used as an augmentation strategy in MDD. The development of new 5-HT1A-R agonists with selectivity for postsynaptic 5-HT1A-Rs may open new perspectives in the field.
Collapse
|
36
|
Du X, Pang TYC, Hannan AJ. A Tale of Two Maladies? Pathogenesis of Depression with and without the Huntington's Disease Gene Mutation. Front Neurol 2013; 4:81. [PMID: 23847583 PMCID: PMC3705171 DOI: 10.3389/fneur.2013.00081] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 06/11/2013] [Indexed: 12/23/2022] Open
Abstract
Huntington’s disease (HD) is an autosomal dominant disorder caused by a tandem repeat expansion encoding an expanded tract of glutamines in the huntingtin protein. HD is progressive and manifests as psychiatric symptoms (including depression), cognitive deficits (culminating in dementia), and motor abnormalities (including chorea). Having reached the twentieth anniversary of the discovery of the “genetic stutter” which causes HD, we still lack sophisticated insight into why so many HD patients exhibit affective disorders such as depression at very early stages, prior to overt appearance of motor deficits. In this review, we will focus on depression as the major psychiatric manifestation of HD, discuss potential mechanisms of pathogenesis identified from animal models, and compare depression in HD patients with that of the wider gene-negative population. The discovery of depressive-like behaviors as well as cellular and molecular correlates of depression in transgenic HD mice has added strong support to the hypothesis that the HD mutation adds significantly to the genetic load for depression. A key question is whether HD-associated depression differs from that in the general population. Whilst preclinical studies, clinical data, and treatment responses suggest striking similarities, there are also some apparent differences. We discuss various molecular and cellular mechanisms which may contribute to depression in HD, and whether they may generalize to other depressive disorders. The autosomal dominant nature of HD and the existence of models with excellent construct validity provide a unique opportunity to understand the pathogenesis of depression and associated gene-environment interactions. Thus, understanding the pathogenesis of depression in HD may not only facilitate tailored therapeutic approaches for HD sufferers, but may also translate to the clinical depression which devastates the lives of so many people.
Collapse
Affiliation(s)
- Xin Du
- Behavioural Neuroscience Division, Florey Institute of Neuroscience and Mental Health, University of Melbourne , Parkville, VIC , Australia
| | | | | |
Collapse
|
37
|
Withdrawal symptoms and rebound syndromes associated with switching and discontinuing atypical antipsychotics: theoretical background and practical recommendations. CNS Drugs 2013; 27:545-72. [PMID: 23821039 DOI: 10.1007/s40263-013-0079-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
With the widespread use of atypical or second-generation antipsychotics, switching treatment has become current practice and more complicated, as the pharmacological profiles of these agents differ substantially despite their similarity in being 'atypical'. All share the ability to block dopamine D₂ receptors, and most of them also block serotonin 5-HT2A receptors. Apart from these common features, some atypical antipsychotics are also able to block or stimulate other dopamine or serotonin receptors, as well as histaminergic, muscarinergic or adrenergic receptors. As a result of the varying receptor affinities, in switching or discontinuing compounds several possible pitfalls have to be considered, including the occurrence of withdrawal and rebound syndromes. This article reviews the pharmacological background of functional blockade or stimulation of receptors of interest in regard to atypical antipsychotics and the implicated potential withdrawal and rebound phenomena. A MEDLINE search was carried out to identify information on withdrawal or rebound syndromes occurring after discontinuation of atypical antipsychotics. Using the resulting literature, we first discuss the theoretical background to the functional consequences of atypical antipsychotic-induced blockade or stimulation of neurotransmitter receptors and, secondly, we highlight the clinical consequences of this. We then review the available clinical literature on switching between atypical antipsychotics, with respect to the occurrence of withdrawal or rebound symptoms. Finally, we offer practical recommendations based on the reviewed findings. The systematic evaluation of withdrawal or rebound phenomena using randomized controlled trials is still understudied. Knowledge of pharmacological receptor-binding profiles may help clinicians in choosing adequate switching or discontinuation strategies for each agent. Results from large switching trials indicate that switching atypical antipsychotics can be performed in a safe manner. Treatment-emergent adverse events during or after switching are not always considered to be, at least in part, associated with the pre-switch antipsychotic. Further studies are needed to substantiate the evidence gained so far on different switching strategies. The use of concomitant medication, e.g., benzodiazepines or anticholinergic drugs, may help to minimize symptoms arising from the discontinuation or switching of antipsychotic treatment.
Collapse
|
38
|
Olivier JDA, Vinkers CH, Olivier B. The role of the serotonergic and GABA system in translational approaches in drug discovery for anxiety disorders. Front Pharmacol 2013; 4:74. [PMID: 23781201 PMCID: PMC3677985 DOI: 10.3389/fphar.2013.00074] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 05/21/2013] [Indexed: 12/13/2022] Open
Abstract
There is ample evidence that genetic factors play an important role in anxiety disorders. In support, human genome-wide association studies have implicated several novel candidate genes. However, illumination of such genetic factors involved in anxiety disorders has not resulted in novel drugs over the past decades. A complicating factor is the heterogeneous classification of anxiety disorders in the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV-TR) and diverging operationalization of anxiety used in preclinical and clinical studies. Currently, there is an increasing focus on the gene × environment (G × E) interaction in anxiety as genes do not operate in isolation and environmental factors have been found to significantly contribute to the development of anxiety disorders in at-risk individuals. Nevertheless, extensive research on G × E mechanisms in anxiety has not resulted in major breakthroughs in drug discovery. Modification of individual genes in rodent models has enabled the specific study of anxiety in preclinical studies. In this context, two extensively studied neurotransmitters involved in anxiety are the gamma-aminobutyric acid (GABA) and 5-HT (5-hydroxytryptamine) system. In this review, we illustrate the complex interplay between genes and environment in anxiety processes by reviewing preclinical and clinical studies on the serotonin transporter (5-HTT), 5-HT1A receptor, 5-HT2 receptor, and GABAA receptor. Even though targets from the serotonin and GABA system have yielded drugs with known anxiolytic efficacy, the relation between the genetic background of these targets and anxiety symptoms and development of anxiety disorders is largely unknown. The aim of this review is to show the vast complexity of genetic and environmental factors in anxiety disorders. In light of the difficulty with which common genetic variants are identified in anxiety disorders, animal models with translational validity may aid in elucidating the neurobiological background of these genes and their possible role in anxiety. We argue that, in addition to human genetic studies, translational models are essential to map anxiety-related genes and to enhance our understanding of anxiety disorders in order to develop potentially novel treatment strategies.
Collapse
Affiliation(s)
- Jocelien D A Olivier
- Department of, Women's and Children's Health, Uppsala University Uppsala, Sweden ; Center for Gender Medicine, Karolinska Institutet Stockholm, Sweden
| | | | | |
Collapse
|
39
|
De Filippis B, Ricceri L, Fuso A, Laviola G. Neonatal exposure to low dose corticosterone persistently modulates hippocampal mineralocorticoid receptor expression and improves locomotor/exploratory behaviour in a mouse model of Rett syndrome. Neuropharmacology 2013; 68:174-83. [DOI: 10.1016/j.neuropharm.2012.05.048] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 04/12/2012] [Accepted: 05/28/2012] [Indexed: 12/20/2022]
|
40
|
Zavitsanou K, Dalton VS, Walker AK, Weickert CS, Sominsky L, Hodgson DM. Neonatal lipopolysaccharide treatment has long-term effects on monoaminergic and cannabinoid receptors in the rat. Synapse 2013; 67:290-9. [DOI: 10.1002/syn.21640] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 01/29/2013] [Indexed: 12/20/2022]
Affiliation(s)
| | - Victoria S. Dalton
- Department of Psychiatry and Trinity College Institute of Neuroscience; Trinity College Dublin; Dublin; Ireland
| | | | | | - Luba Sominsky
- Laboratory of Neuroimmunology, School of Psychology; University of Newcastle; Newcastle; Australia
| | - Deborah M. Hodgson
- Laboratory of Neuroimmunology, School of Psychology; University of Newcastle; Newcastle; Australia
| |
Collapse
|
41
|
Altieri SC, Garcia-Garcia AL, Leonardo ED, Andrews AM. Rethinking 5-HT1A receptors: emerging modes of inhibitory feedback of relevance to emotion-related behavior. ACS Chem Neurosci 2013; 4:72-83. [PMID: 23336046 DOI: 10.1021/cn3002174] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 12/20/2012] [Indexed: 11/27/2022] Open
Abstract
The complexities of the involvement of the serotonin transmitter system in numerous biological processes and psychiatric disorders is, to a substantial degree, attributable to the large number of serotonin receptor families and subtypes that have been identified and characterized for over four decades. Of these, the 5-HT(1A) receptor subtype, which was the first to be cloned and characterized, has received considerable attention based on its purported role in the etiology and treatment of mood and anxiety disorders. 5-HT(1A) receptors function both at presynaptic (autoreceptor) and postsynaptic (heteroreceptor) sites. Recent research has implicated distinct roles for these two populations of receptors in mediating emotion-related behavior. New concepts as to how 5-HT(1A) receptors function to control serotonergic tone throughout life were highlights of the proceedings of the 2012 Serotonin Club Meeting in Montpellier, France. Here, we review recent findings and current perspectives on functional aspects of 5-HT(1A) auto- and heteroreceptors with particular regard to their involvement in altered anxiety and mood states.
Collapse
Affiliation(s)
| | - Alvaro L. Garcia-Garcia
- Department of Psychiatry, Division of Integrative Neuroscience, Columbia University and New York State Psychiatric Institute, New York, New York 10032, United States
| | - E. David Leonardo
- Department of Psychiatry, Division of Integrative Neuroscience, Columbia University and New York State Psychiatric Institute, New York, New York 10032, United States
| | | |
Collapse
|
42
|
Albert PR. Transcriptional regulation of the 5-HT1A receptor: implications for mental illness. Philos Trans R Soc Lond B Biol Sci 2012; 367:2402-15. [PMID: 22826341 DOI: 10.1098/rstb.2011.0376] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The serotonin-1A (5-HT(1A)) receptor is an abundant post-synaptic 5-HT receptor (heteroreceptor) implicated in regulation of mood, emotion and stress responses and is the major somatodendritic autoreceptor that negatively regulates 5-HT neuronal activity. Based on animal models, an integrated model for opposing roles of pre- and post-synaptic 5-HT(1A) receptors in anxiety and depression phenotypes and response to antidepressants is proposed. Understanding differential transcriptional regulation of pre- versus post-synaptic 5-HT(1A) receptors could provide better tools for their selective regulation. This review examines the transcription factors that regulate brain region-specific basal and stress-induced expression of the 5-HT(1A) receptor gene (Htr1a). A functional polymorphism, rs6295 in the Htr1a promoter region, blocks the function of specific repressors Hes1, Hes5 and Deaf1, resulting in increased 5-HT(1A) autoreceptor expression in animal models and humans. Its association with altered 5-HT(1A) expression, depression, anxiety and antidepressant response are related to genotype frequency in different populations, sample homogeneity, disease outcome measures and severity. Preliminary evidence from gene × environment studies suggests the potential for synergistic interaction of stress-mediated repression of 5-HT(1A) heteroreceptors, and rs6295-induced upregulation of 5-HT(1A) autoreceptors. Targeted therapeutics to inhibit 5-HT(1A) autoreceptor expression and induce 5-HT(1A) heteroreceptor expression may ameliorate treatment of anxiety and major depression.
Collapse
Affiliation(s)
- Paul R Albert
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa, , 451 Smyth Road, Ottawa, ON, Canada , K1H 8M5.
| |
Collapse
|
43
|
Ricci LA, Morrison TR, Melloni RH. Serotonin modulates anxiety-like behaviors during withdrawal from adolescent anabolic-androgenic steroid exposure in Syrian hamsters. Horm Behav 2012; 62:569-78. [PMID: 23026540 PMCID: PMC3612524 DOI: 10.1016/j.yhbeh.2012.09.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 09/20/2012] [Accepted: 09/21/2012] [Indexed: 12/27/2022]
Abstract
From the U.S. to Europe and Australia anabolic steroid abuse remains high in the adolescent population. This is concerning given that anabolic steroid use is associated with a higher incidence of pathological anxiety that often appears during withdrawal from use. This study uses pubertal Syrian hamsters (Mesocricetus auratus) to investigate the hypothesis that adolescent anabolic/androgenic steroid (AAS) exposure predisposes hamsters to heightened levels of anxiety during AAS withdrawal that is modulated by serotonin (5HT) neural signaling. In the first two sets of experiments, adolescent AAS-treated hamsters were tested for anxiety 21 days after the cessation of AAS administration (i.e., during AAS withdrawal) using the elevated plus maze (EPM), dark/light (DL), and seed finding (SF) tests and then examined for differences in 5HT afferent innervation to select areas of the brain important for anxiety. In the EPM and DL tests, adolescent AAS exposure leads to significant increases in anxiety-like response during AAS withdrawal. AAS-treated hamsters showed long-term reductions in 5HT innervation within several areas of the hamster brain implicated in anxiety, most notably the anterior hypothalamus and the central and medial amygdala. However, no differences in 5HT were found in other anxiety areas, e.g., frontal cortex and lateral septum. In the last experiment, adolescent AAS-treated hamsters were scored for anxiety on the 21st day of AAS withdrawal following the systemic administration of saline or one of three doses of fluoxetine, a selective serotonin reuptake inhibitor. Saline-treated hamsters showed high levels of AAS withdrawal-induced anxiety, while treatment with fluoxetine reduced AAS withdrawal-induced anxiety. These findings indicate that early AAS exposure has potent anxiogenic effects during AAS withdrawal that are modulated, in part, by 5HT signaling.
Collapse
|
44
|
Early intervention with fluoxetine reverses abnormalities in the serotonergic system and behavior of rats exposed prenatally to dexamethasone. Neuropharmacology 2012; 63:292-300. [DOI: 10.1016/j.neuropharm.2012.03.027] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 03/29/2012] [Accepted: 03/31/2012] [Indexed: 11/19/2022]
|
45
|
Repeated exposures to chlorpyrifos lead to spatial memory retrieval impairment and motor activity alteration. Neurotoxicol Teratol 2012; 34:442-9. [PMID: 22640976 DOI: 10.1016/j.ntt.2012.05.053] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2011] [Revised: 05/09/2012] [Accepted: 05/15/2012] [Indexed: 11/22/2022]
Abstract
Chlorpyrifos (CPF) is one of the most commonly used insecticides throughout the world and has become one of the major pesticides detected in farm products. Chronic exposures to CPF, especially at the dosages without eliciting any systemic toxicity, require greater attention. The purpose of this study was, therefore, to evaluate the behavioral effects of repeated low doses (doses that do not produce overt signs of cholinergic toxicity) of CPF in adult rats. Male rats were given 0, 1.0, 5.0 or 10.0mg/kg of CPF through intragastric administration daily for 4 consecutive weeks. The behavioral functions were assessed in a series of behavioral tests, including water maze task, open-field test, grip strength and rotarod test. Furthermore, the present study was designed to evaluate the effects of repeated exposures to CPF on water maze recall and not acquisition. The results showed that the selected doses only had mild inhibition effects on cholinesterase activity, and have no effects on weight gain and daily food consumption. Performances in the spatial retention task (Morris water maze) were impaired after the 4-week exposure to CPF, but the performances of grip strength and rotarod test were not affected. Motor activities in the open field were changed, especially the time spent in the central zone increased. The results indicated that repeated exposures to low doses of CPF may lead to spatial recall impairments, behavioral abnormalities. However, the underlying mechanism needs further investigations.
Collapse
|
46
|
Effects of environmental manipulations in genetically targeted animal models of affective disorders. Neurobiol Dis 2012; 57:12-27. [PMID: 22525570 DOI: 10.1016/j.nbd.2012.04.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 04/02/2012] [Accepted: 04/06/2012] [Indexed: 12/31/2022] Open
Abstract
Mental illness is the leading cause of disability worldwide. We are only just beginning to reveal and comprehend the complex interaction that exists between the genetic makeup of an organism and the potential modifying effect of the environment in which it lives, and how this translates into mediating susceptibility to neurological and psychiatric conditions. The capacity to address this issue experimentally has been facilitated by the availability of rodent models which allow the precise manipulation of genetic and environmental factors. In this review, we discuss the valuable nature of animal models in furthering our understanding of the relationship between genetic and environmental factors in affective illnesses, such as anxiety and depressive disorders. We first highlight the behavioral impairments exhibited by genetically targeted animal models of affective disorders, and then provide a discussion of the underlying neurobiology, focusing on animal models that involve exposure to stress. This is followed by a review of recent studies that report of beneficial effects of environmental manipulations such as environmental enrichment and enhanced physical activity and discuss the likely mechanisms that mediate those benefits.
Collapse
|
47
|
Czesak M, Le François B, Millar AM, Deria M, Daigle M, Visvader JE, Anisman H, Albert PR. Increased serotonin-1A (5-HT1A) autoreceptor expression and reduced raphe serotonin levels in deformed epidermal autoregulatory factor-1 (Deaf-1) gene knock-out mice. J Biol Chem 2012; 287:6615-27. [PMID: 22232550 PMCID: PMC3307310 DOI: 10.1074/jbc.m111.293027] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 12/23/2011] [Indexed: 02/02/2023] Open
Abstract
Altered regulation of the serotonin-1A (5-HT1A) receptor gene is implicated in major depression and mood disorders. The functional human 5-HT1A C(-1019)G promoter polymorphism (rs6295), which prevents the binding of Deaf-1/NUDR leading to dysregulation of the receptor, has been associated with major depression. In cell models Deaf-1 displays dual activity, repressing 5-HT1A autoreceptor expression in serotonergic raphe cells while enhancing postsynaptic 5-HT1A heteroreceptor expression in nonserotonergic neurons. A functional Deaf-1 binding site on the mouse 5-HT1A promoter was recognized by Deaf-1 in vitro and in vivo and mediated dual activity of Deaf-1 on 5-HT1A gene transcription. To address regulation by Deaf-1 in vivo, Deaf-1 knock-out mice bred to a C57BL/6 background were compared with wild-type siblings for changes in 5-HT1A RNA and protein by quantitative RT-PCR, in situ hybridization, and immunofluorescence. In the dorsal raphe, Deaf-1 knock-out mice displayed increased 5-HT1A mRNA, protein, and 5-HT1A-positive cell counts but reduced 5-HT levels, whereas other serotonergic markers, such as tryptophan hydroxylase (TPH)- or 5-HT-positive cells and TPH2 RNA levels, were unchanged. By contrast, 5-HT1A mRNA and 5-HT1A-positive cells were reduced in the frontal cortex of Deaf-1-null mice, with no significant change in hippocampal 5-HT1A RNA, protein, or cell counts. The region-specific alterations of brain 5-HT1A gene expression and reduced raphe 5-HT content in Deaf-1(-/-) mice indicate the importance of Deaf-1 in regulation of 5-HT1A gene expression and provide insight into the role of the 5-HT1A G(-1019) allele in reducing serotonergic neurotransmission by derepression of 5-HT1A autoreceptors.
Collapse
MESH Headings
- Animals
- Autoreceptors/genetics
- Autoreceptors/metabolism
- DNA-Binding Proteins
- Depressive Disorder/metabolism
- Depressive Disorder/physiopathology
- Female
- Fluorescent Antibody Technique
- Male
- Mice
- Mice, 129 Strain
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Polymorphism, Genetic/genetics
- Promoter Regions, Genetic/genetics
- RNA, Messenger/metabolism
- Raphe Nuclei/physiology
- Receptor, Serotonin, 5-HT1A/genetics
- Receptor, Serotonin, 5-HT1A/metabolism
- Serotonin/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Tryptophan Hydroxylase/genetics
- Tryptophan Hydroxylase/metabolism
Collapse
Affiliation(s)
- Margaret Czesak
- From the Ottawa Hospital Research Institute (Neuroscience), Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Brice Le François
- From the Ottawa Hospital Research Institute (Neuroscience), Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Anne M. Millar
- From the Ottawa Hospital Research Institute (Neuroscience), Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Mariam Deria
- From the Ottawa Hospital Research Institute (Neuroscience), Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Mireille Daigle
- From the Ottawa Hospital Research Institute (Neuroscience), Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Jane E. Visvader
- the Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3050, Australia, and
| | - Hymie Anisman
- the Institute of Neuroscience, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | - Paul R. Albert
- From the Ottawa Hospital Research Institute (Neuroscience), Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
48
|
Fournet V, Schweitzer A, Chevarin C, Deloulme JC, Hamon M, Giros B, Andrieux A, Martres MP. The deletion of STOP/MAP6 protein in mice triggers highly altered mood and impaired cognitive performances. J Neurochem 2012; 121:99-114. [PMID: 22146001 DOI: 10.1111/j.1471-4159.2011.07615.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The microtubule-associated Stable Tubulie Only Polypeptide (STOP; also known as MAP6) protein plays a key role in neuron architecture and synaptic plasticity, the dysfunctions of which are thought to be implicated in the pathophysiology of psychiatric diseases. The deletion of STOP in mice leads to severe disorders reminiscent of several schizophrenia-like symptoms, which are also associated with differential alterations of the serotonergic tone in somas versus terminals. In STOP knockout (KO) compared with wild-type mice, serotonin (5-HT) markers are found to be markedly accumulated in the raphe nuclei and, in contrast, deeply depleted in all serotonergic projection areas. In the present study, we carefully examined whether the 5-HT imbalance would lead to behavioral consequences evocative of mood and/or cognitive disorders. We showed that STOP KO mice exhibited depression-like behavior, associated with a decreased anxiety-status in validated paradigms. In addition, although STOP KO mice had a preserved very short-term memory, they failed to perform well in all other learning and memory tasks. We also showed that STOP KO mice exhibited regional imbalance of the norepinephrine tone as observed for 5-HT. As a consequence, mutant mice were hypersensitive to acute antidepressants with different selectivity. Altogether, these data indicate that the deletion of STOP protein in mice caused deep alterations in mood and cognitive performances and that STOP protein might have a crucial role in the 5-HT and norepinephrine networks development.
Collapse
Affiliation(s)
- Vincent Fournet
- INSERM UMRS 952, CNRS UMR 7224, Université Pierre et Marie Curie, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Shrestha S, Hirvonen J, Hines CS, Henter ID, Svenningsson P, Pike VW, Innis RB. Serotonin-1A receptors in major depression quantified using PET: controversies, confounds, and recommendations. Neuroimage 2011; 59:3243-51. [PMID: 22155042 DOI: 10.1016/j.neuroimage.2011.11.029] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 11/05/2011] [Accepted: 11/09/2011] [Indexed: 01/21/2023] Open
Abstract
The serotonin-1A (5-HT(1A)) receptor is of particular interest in human positron emission tomography (PET) studies of major depressive disorder (MDD). Of the eight studies investigating this issue in the brains of patients with MDD, four reported decreased 5-HT(1A) receptor density, two reported no change, and two reported increased 5-HT(1A) receptor density. While clinical heterogeneity may have contributed to these differing results, methodological factors by themselves could also explain the discrepancies. This review highlights several of these factors, including the use of the cerebellum as a reference region and the imprecision of measuring the concentration of parent radioligand in arterial plasma, the method otherwise considered to be the 'gold standard'. Other potential confounds also exist that could restrict or unexpectedly affect the interpretation of results. For example, the radioligand may be a substrate for an efflux transporter - like P-gp - at the blood-brain barrier; furthermore, the binding of the radioligand to the receptor in various stages of cellular trafficking is unknown. Efflux transport and cellular trafficking may also be differentially expressed in patients compared to healthy subjects. We believe that, taken together, the existing disparate findings do not reliably answer the question of whether 5-HT(1A) receptors are altered in MDD or in subgroups of patients with MDD. In addition, useful meta-analysis is precluded because only one of the imaging centers acquired all the data necessary to address these methodological concerns. We recommend that in the future, individual centers acquire more thorough data capable of addressing methodological concerns, and that multiple centers collaborate to meaningfully pool their data for meta-analysis.
Collapse
Affiliation(s)
- Saurav Shrestha
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, MSC 1026, Bldg 10, Room B1D43, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
50
|
A role for 5-HT1A receptors in the basolateral amygdala in the development of conditioned defeat in Syrian hamsters. Pharmacol Biochem Behav 2011; 100:592-600. [PMID: 21967885 DOI: 10.1016/j.pbb.2011.09.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 09/12/2011] [Accepted: 09/16/2011] [Indexed: 11/22/2022]
Abstract
The basolateral nucleus of the amygdala (BLA) is a key brain region regulating behavioral changes following stressful events, including social defeat. Previous research has shown that activation of serotonin (5-HT) 1A receptors in the BLA reduces conditioned fear and anxiety-like behavior. The objective of this study was to test whether 5-HT1A receptors in the BLA contribute to conditioned defeat in male Syrian hamsters (Mesocricetus auratus). We tested whether injection of the selective 5-HT1A receptor agonist flesinoxan (400 ng, 800 ng, or 1200 ng in 200 nl saline) into the BLA prior to social defeat would reduce the acquisition of conditioned defeat, and whether a similar injection prior to testing would reduce the expression of conditioned defeat. We also tested whether injection of the selective 5-HT1A receptor antagonist WAY-100635 (400 ng or 1600 ng in 200 nl saline) into the BLA prior to social defeat would enhance the acquisition of conditioned defeat, and whether a similar injection prior to testing would enhance the expression of conditioned defeat. We found that injection of flesinoxan into the BLA decreased both the acquisition and expression of conditioned defeat. However, injection of WAY-100635 into the BLA did not alter the acquisition or expression of conditioned defeat. These data indicate that pharmacological activation of 5-HT1A receptors in the BLA is sufficient to impair the acquisition and expression of conditioned defeat. Our results suggest that pharmacological treatments that activate 5-HT1A receptors in the BLA are capable of reducing the development of stress-induced changes in behavior.
Collapse
|