1
|
Parkin C, Ortiz J, Cruz S, Bath KG, Romeo RD. Pubertal- and Stress-Dependent Changes in Cellular Activation and Expression of Excitatory Amino Acid Receptor Subunits in the Paraventricular Nucleus of the Hypothalamus in Male and Female Rats. Dev Neurosci 2024:1-11. [PMID: 39467516 PMCID: PMC12034827 DOI: 10.1159/000542277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/23/2024] [Indexed: 10/30/2024] Open
Abstract
INTRODUCTION Pubertal maturation is marked by significant changes in stress-induced hormonal responses mediated by the hypothalamic-pituitary-adrenal (HPA) axis, with prepubertal male and female rats often exhibiting greater HPA reactivity compared to adult males and females. Though the implications of these changes are unclear, elevated stress responsiveness might contribute to the stress-related vulnerabilities often associated with puberty. METHODS The current experiments sought to determine whether differences in cellular activation, as measured by FOS immunohistochemistry, or excitatory ionotropic glutamate receptor subunit expression, as measured by qRT-PCR, in the paraventricular nucleus (PVN) were associated with these noted pubertal shifts in stress reactivity in male and female rats. As the PVN is the key nucleus responsible for activating the hormonal stress response, we predicted greater cellular activation and higher expression levels of glutamate receptor subunits in the PVN of prepubertal males and females compared to their adult counterparts. RESULTS Our FOS data revealed that while prepubertal males showed greater stress-induced activation in the PVN than adult males, prepubertal females showed less activation than adult females. Moreover, many of the NMDA, AMPA, and kainate receptor subunits measured, including Grin1, Grin2b, Gria1, Gria2, Grik1, and Grik2, had higher expression levels in adults, particularly in males. CONCLUSIONS Though not supporting our initial predictions, these data do indicate that age and stress influence the activation of the PVN and the expression of glutamate receptor subunits important in its function. These data also suggest that the effects of age and stress are different in males and females. Though still far from a clear understanding of what mechanism(s) mediate pubertal shift in stress reactivity, these data add to our growing understanding of how age, stress, and sex influence HPA function.
Collapse
Affiliation(s)
- Catherine Parkin
- Barnard College of Columbia University, Department of Neuroscience and Behavior New York, NY
| | - Juliet Ortiz
- Barnard College of Columbia University, Department of Neuroscience and Behavior New York, NY
| | - Sofia Cruz
- Barnard College of Columbia University, Department of Neuroscience and Behavior New York, NY
| | - Kevin G. Bath
- Columbia University, Department of Psychiatry, New York, NY
- New York State Psychiatric Institute, Division of Developmental Neuroscience, New York, NY
| | - Russell D. Romeo
- Barnard College of Columbia University, Department of Neuroscience and Behavior New York, NY
| |
Collapse
|
2
|
Cao K, Zhong J, Wang S, Shi Y, Bai S, Zhao J, Yang L, Liang Q, Deng D, Zhang R. SiNiSan exerts antidepressant effects by modulating serotonergic/GABAergic neuron activity in the dorsal raphe nucleus region through NMDA receptor in the adolescent depression mouse model. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118040. [PMID: 38479542 DOI: 10.1016/j.jep.2024.118040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/29/2024] [Accepted: 03/09/2024] [Indexed: 03/25/2024]
Affiliation(s)
- Kerun Cao
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jialong Zhong
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shanshan Wang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yafei Shi
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shasha Bai
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinlan Zhao
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Yang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Liang
- Shenzhen Bao'an Traditional Chinese Medicine Hospital Group, Shenzhen, 518000, China.
| | - Di Deng
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Rong Zhang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
3
|
Flynn LT, Gao WJ. DNA methylation and the opposing NMDAR dysfunction in schizophrenia and major depression disorders: a converging model for the therapeutic effects of psychedelic compounds in the treatment of psychiatric illness. Mol Psychiatry 2023; 28:4553-4567. [PMID: 37679470 PMCID: PMC11034997 DOI: 10.1038/s41380-023-02235-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/09/2023]
Abstract
Psychedelic compounds are being increasingly explored as a potential therapeutic option for treating several psychiatric conditions, despite relatively little being known about their mechanism of action. One such possible mechanism, DNA methylation, is a process of epigenetic regulation that changes gene expression via chemical modification of nitrogenous bases. DNA methylation has been implicated in the pathophysiology of several psychiatric conditions, including schizophrenia (SZ) and major depressive disorder (MDD). In this review, we propose alterations to DNA methylation as a converging model for the therapeutic effects of psychedelic compounds, highlighting the N-methyl D-aspartate receptor (NMDAR), a crucial mediator of synaptic plasticity with known dysfunction in both diseases, as an example and anchoring point. We review the established evidence relating aberrant DNA methylation to NMDAR dysfunction in SZ and MDD and provide a model asserting that psychedelic substances may act through an epigenetic mechanism to provide therapeutic effects in the context of these disorders.
Collapse
Affiliation(s)
- L Taylor Flynn
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
- MD/PhD program, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Wen-Jun Gao
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Chen B, Xu J, Chen S, Mou T, Wang Y, Wang H, Zhang Z, Ren F, Wang Z, Jin K, Lu J. Dysregulation of striatal dopamine D2/D3 receptor-mediated by hypocretin induces depressive behaviors in rats. J Affect Disord 2023; 325:256-263. [PMID: 36638964 DOI: 10.1016/j.jad.2023.01.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/26/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
BACKGROUND The dysregulation of the dopamine system contributes to depressive-like behaviors in rats, and the neurological functions regulated by hypocretin are severely affected in depression. However, whether suvorexant plays a role in alleviating depression by affecting the dopamine system is unclear. METHODS To preliminarily explore the mechanism of suvorexant (10 mg/kg) in the treatment of depression, the mRNA and protein expression of TH, Drd2, Drd3, GluN2A, DAT, and GluN2B in the striatum of rats was quantified by qPCR and western blotting. The plasma hypocretin-1 and dopamine levels and the striatal dopamine levels were determined by ELISA. RESULTS i) Compared to those of the control group, chronic unpredictable mild stress (CUMS) rats showed depressive-like behaviors, which were subsequently reversed by treatment with suvorexant. ii) The mRNA and protein expressions of TH, Drd2, Drd3, GluN2A, and GluN2B in the striatum of CUMS were significantly increased compared with those in the controls, but decreased after suvorexant treatment. iii) Compared with those in the control group, the plasma and striatal dopamine levels of CUMS decreased while plasma hypocretin-1 levels increased, which was reversed after suvorexant treatment. LIMITATIONS i) The suvorexant is a dual hypocretin receptor antagonist; however, the responsible receptor is unclear. ii) We only focused on related factors in the striatum but did not explore other brain regions, nor did we directly explore the relationship among these factors. CONCLUSION Depressive-like behaviors induced by CUMS can be reversed by suvorexant, and the therapeutic effects of suvorexant may be mediated by affecting the dopamine system.
Collapse
Affiliation(s)
- Bing Chen
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Jiangang Xu
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999007, Hong Kong
| | - Simiao Chen
- School of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China
| | - Tingting Mou
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Ying Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Haojun Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhihan Zhang
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Feifan Ren
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Zheng Wang
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Kangyu Jin
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China.
| | - Jing Lu
- Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China.
| |
Collapse
|
5
|
Demin KA, Krotova NA, Ilyin NP, Galstyan DS, Kolesnikova TO, Strekalova T, de Abreu MS, Petersen EV, Zabegalov KN, Kalueff AV. Evolutionarily conserved gene expression patterns for affective disorders revealed using cross-species brain transcriptomic analyses in humans, rats and zebrafish. Sci Rep 2022; 12:20836. [PMID: 36460699 PMCID: PMC9718822 DOI: 10.1038/s41598-022-22688-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/18/2022] [Indexed: 12/03/2022] Open
Abstract
Widespread, debilitating and often treatment-resistant, depression and other stress-related neuropsychiatric disorders represent an urgent unmet biomedical and societal problem. Although animal models of these disorders are commonly used to study stress pathogenesis, they are often difficult to translate across species into valuable and meaningful clinically relevant data. To address this problem, here we utilized several cross-species/cross-taxon approaches to identify potential evolutionarily conserved differentially expressed genes and their sets. We also assessed enrichment of these genes for transcription factors DNA-binding sites down- and up- stream from their genetic sequences. For this, we compared our own RNA-seq brain transcriptomic data obtained from chronically stressed rats and zebrafish with publicly available human transcriptomic data for patients with major depression and their respective healthy control groups. Utilizing these data from the three species, we next analyzed their differential gene expression, gene set enrichment and protein-protein interaction networks, combined with validated tools for data pooling. This approach allowed us to identify several key brain proteins (GRIA1, DLG1, CDH1, THRB, PLCG2, NGEF, IKZF1 and FEZF2) as promising, evolutionarily conserved and shared affective 'hub' protein targets, as well as to propose a novel gene set that may be used to further study affective pathogenesis. Overall, these approaches may advance cross-species brain transcriptomic analyses, and call for further cross-species studies into putative shared molecular mechanisms of affective pathogenesis.
Collapse
Affiliation(s)
- Konstantin A Demin
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia.
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia.
| | - Nataliya A Krotova
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Nikita P Ilyin
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia
| | - David S Galstyan
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | | | | | | | | | | | - Allan V Kalueff
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia.
- Institute of Neurosciences and Medicine, Novosibirsk, Russia.
- Ural Federal University, Ekaterinburg, Russia.
| |
Collapse
|
6
|
Hrivikova K, Zelena D, Graban J, Puhova A, Miklya I, Balazsfi D, Jezova D. Chronic treatment with enhancer drugs modifies the gene expression of selected parameters related to brain plasticity in rats under stress conditions. Neurochem Int 2022; 159:105404. [PMID: 35853552 DOI: 10.1016/j.neuint.2022.105404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 07/08/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022]
Abstract
Selegiline, also known as L-deprenyl, and (2R)-1-(1-benzofuran-2-yl)-N-propylpentane-2-amine (BPAP) were found to induce enhancement of monoamine neurotransmission in low and very low doses. In addition, these enhancers may modify glutamatergic neurotransmission. The aim of the present study was to test the hypothesis that under stress conditions, chronic treatment with enhancer drugs has a positive impact on the glutamatergic system and other parameters related to brain plasticity, stress-related systems, and anxiety behavior. We exposed male Wistar rats to a chronic mild stress procedure combined with chronic treatment with two synthetic enhancer drugs. The gene expression of GluR1, an AMPA receptor subunit was reduced by repeated treatment with deprenyl in the hippocampus and with both BPAP and deprenyl in the prefrontal cortex. A significant reduction of NMDA receptor subunit GluN2B expression was observed in the hippocampus but not in the prefrontal cortex. Deprenyl treatment led to an enhancement of hippocampal BDNFmRNA concentrations in stress-exposed rats. Treatment with enhancer drugs failed to induce significant changes in stress hormone concentrations or anxiety behavior. In conclusion, the present study in chronically stressed rats showed that concomitant treatment with enhancer drugs did not provoke substantial neuroendocrine changes, but modified gene expression of selected parameters associated with brain plasticity. Observed changes may indicate a positive influence of enhancer drugs on brain plasticity, which is important for preventing negative consequences of chronic stress and enhancement of stress resilience. It may be suggested that the changes in glutamate receptor subunits induced by enhancer drugs are brain region-specific and not dose-related.
Collapse
Affiliation(s)
- K Hrivikova
- Laboratory of Pharmacological Neuroendocrinology, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - D Zelena
- Institute of Physiology, Medical School, University of Pécs, Centre for Neuroscience, Szentágothai Research Centre, 7624, Pécs, Hungary; Department of Behavioral Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Szigony 43, Hungary
| | - J Graban
- Laboratory of Pharmacological Neuroendocrinology, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - A Puhova
- Laboratory of Pharmacological Neuroendocrinology, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - I Miklya
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089, Budapest, Nagyvarad Ter 4, Hungary
| | - D Balazsfi
- Department of Behavioral Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 1083, Budapest, Szigony 43, Hungary
| | - D Jezova
- Laboratory of Pharmacological Neuroendocrinology, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia.
| |
Collapse
|
7
|
Royo M, Escolano BA, Madrigal MP, Jurado S. AMPA Receptor Function in Hypothalamic Synapses. Front Synaptic Neurosci 2022; 14:833449. [PMID: 35173598 PMCID: PMC8842481 DOI: 10.3389/fnsyn.2022.833449] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 01/03/2022] [Indexed: 12/15/2022] Open
Abstract
AMPA receptors (AMPARs) are critical for mediating glutamatergic synaptic transmission and plasticity, thus playing a major role in the molecular machinery underlying cellular substrates of memory and learning. Their expression pattern, transport and regulatory mechanisms have been extensively studied in the hippocampus, but their functional properties in other brain regions remain poorly understood. Interestingly, electrophysiological and molecular evidence has confirmed a prominent role of AMPARs in the regulation of hypothalamic function. This review summarizes the existing evidence on AMPAR-mediated transmission in the hypothalamus, where they are believed to orchestrate the role of glutamatergic transmission in autonomous, neuroendocrine function, body homeostasis, and social behavior.
Collapse
|
8
|
Lori A, Schultebraucks K, Galatzer-Levy I, Daskalakis NP, Katrinli S, Smith AK, Myers AJ, Richholt R, Huentelman M, Guffanti G, Wuchty S, Gould F, Harvey PD, Nemeroff CB, Jovanovic T, Gerasimov ES, Maples-Keller JL, Stevens JS, Michopoulos V, Rothbaum BO, Wingo AP, Ressler KJ. Transcriptome-wide association study of post-trauma symptom trajectories identified GRIN3B as a potential biomarker for PTSD development. Neuropsychopharmacology 2021; 46:1811-1820. [PMID: 34188182 PMCID: PMC8357796 DOI: 10.1038/s41386-021-01073-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/26/2021] [Accepted: 05/24/2021] [Indexed: 12/11/2022]
Abstract
Biomarkers that predict symptom trajectories after trauma can facilitate early detection or intervention for posttraumatic stress disorder (PTSD) and may also advance our understanding of its biology. Here, we aimed to identify trajectory-based biomarkers using blood transcriptomes collected in the immediate aftermath of trauma exposure. Participants were recruited from an Emergency Department in the immediate aftermath of trauma exposure and assessed for PTSD symptoms at baseline, 1, 3, 6, and 12 months. Three empirical symptom trajectories (chronic-PTSD, remitting, and resilient) were identified in 377 individuals based on longitudinal symptoms across four data points (1, 3, 6, and 12 months), using latent growth mixture modeling. Blood transcriptomes were examined for association with longitudinal symptom trajectories, followed by expression quantitative trait locus analysis. GRIN3B and AMOTL1 blood mRNA levels were associated with chronic vs. resilient post-trauma symptom trajectories at a transcriptome-wide significant level (N = 153, FDR-corrected p value = 0.0063 and 0.0253, respectively). We identified four genetic variants that regulate mRNA blood expression levels of GRIN3B. Among these, GRIN3B rs10401454 was associated with PTSD in an independent dataset (N = 3521, p = 0.04). Examination of the BrainCloud and GTEx databases revealed that rs10401454 was associated with brain mRNA expression levels of GRIN3B. While further replication and validation studies are needed, our data suggest that GRIN3B, a glutamate ionotropic receptor NMDA type subunit-3B, may be involved in the manifestation of PTSD. In addition, the blood mRNA level of GRIN3B may be a promising early biomarker for the PTSD manifestation and development.
Collapse
Affiliation(s)
- Adriana Lori
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Katharina Schultebraucks
- Department of Emergency Medicine, Columbia University Medical Center, New York, NY, USA
- Data Science Institute, Columbia University, New York, NY, USA
| | - Isaac Galatzer-Levy
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Nikolaos P Daskalakis
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, USA
| | - Seyma Katrinli
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
| | - Alicia K Smith
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
| | - Amanda J Myers
- Department of Psychiatry and Behavioral Sciences, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Ryan Richholt
- Neurogenomics Division and Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Matthew Huentelman
- Neurogenomics Division and Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Guia Guffanti
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, USA
| | - Stefan Wuchty
- Department of Biology, University of Miami, Coral Gables, FL, USA
- Department of Computer Science, University of Miami, Coral Gables, FL, USA
- Institute of Data Science and Computing, University of Miami, Coral Gables, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Felicia Gould
- Department of Psychiatry and Behavioral Sciences, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Philip D Harvey
- Department of Psychiatry and Behavioral Sciences, University of Miami, Miller School of Medicine, Miami, FL, USA
| | | | - Tanja Jovanovic
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University, Detroit, MI, USA
| | | | | | - Jennifer S Stevens
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Vasiliki Michopoulos
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Barbara O Rothbaum
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Aliza P Wingo
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA.
- Division of Mental Health, Atlanta VA Medical Center, Decatur, GA, USA.
| | - Kerry J Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA.
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
9
|
Sales AJ, Maciel IS, Suavinha ACDR, Joca SRL. Modulation of DNA Methylation and Gene Expression in Rodent Cortical Neuroplasticity Pathways Exerts Rapid Antidepressant-Like Effects. Mol Neurobiol 2021; 58:777-794. [PMID: 33025509 DOI: 10.1007/s12035-020-02145-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Stress increases DNA methylation, primarily a suppressive epigenetic mechanism catalyzed by DNA methyltransferases (DNMT), and decreases the expression of genes involved in neuronal plasticity and mood regulation. Despite chronic antidepressant treatment decreases stress-induced DNA methylation, it is not known whether inhibition of DNMT would convey rapid antidepressant-like effects. AIM This work tested such a hypothesis and evaluated whether a behavioral effect induced by DNMT inhibitors (DNMTi) corresponds with changes in DNA methylation and transcript levels in genes consistently associated with the neurobiology of depression and synaptic plasticity (BDNF, TrkB, 5-HT1A, NMDA, and AMPA). METHODS Male Wistar rats received intraperitoneal (i.p.) injection of two pharmacologically different DNMTi (5-AzaD 0.2 and 0.6 mg/kg or RG108 0.6 mg/kg) or vehicle (1 ml/kg), 1 h or 7 days before the learned helplessness test (LH). DNA methylation in target genes and the correspondent transcript levels were measured in the hippocampus (HPC) and prefrontal cortex (PFC) using meDIP-qPCR. In parallel separate groups, the antidepressant-like effect of 5-AzaD and RG108 was investigated in the forced swimming test (FST). The involvement of cortical BDNF-TrkB-mTOR pathways was assessed by intra-ventral medial PFC (vmPFC) injections of rapamycin (mTOR inhibitor), K252a (TrkB receptor antagonist), or vehicle (0.2 μl/side). RESULTS We found that both 5-AzaD and RG108 acutely and 7 days before the test decreased escape failures in the LH. LH stress increased DNA methylation and decreased transcript levels of BDNF IV and TrkB in the PFC, effects that were not significantly attenuated by RG108 treatment. The systemic administration of 5-AzaD (0.2 mg/kg) and RG108 (0.2 mg/kg) induced an antidepressant-like effect in FST, which was, however, attenuated by TrkB and mTOR inhibition into the vmPFC. CONCLUSION These findings suggest that acute inhibition of stress-induced DNA methylation promotes rapid and sustained antidepressant effects associated with increased BDNF-TrkB-mTOR signaling in the PFC.
Collapse
Affiliation(s)
- Amanda J Sales
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
- FMRP-USP, Av Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil.
| | - Izaque S Maciel
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Angélica C D R Suavinha
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Sâmia R L Joca
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
- FCFRP-USP, Av Café, sn, Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil.
| |
Collapse
|
10
|
Candidate Strategies for Development of a Rapid-Acting Antidepressant Class That Does Not Result in Neuropsychiatric Adverse Effects: Prevention of Ketamine-Induced Neuropsychiatric Adverse Reactions. Int J Mol Sci 2020; 21:ijms21217951. [PMID: 33114753 PMCID: PMC7662754 DOI: 10.3390/ijms21217951] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/19/2020] [Accepted: 10/23/2020] [Indexed: 02/08/2023] Open
Abstract
Non-competitive N-methyl-D-aspartate/glutamate receptor (NMDAR) antagonism has been considered to play important roles in the pathophysiology of schizophrenia. In spite of severe neuropsychiatric adverse effects, esketamine (racemic enantiomer of ketamine) has been approved for the treatment of conventional monoaminergic antidepressant-resistant depression. Furthermore, ketamine improves anhedonia, suicidal ideation and bipolar depression, for which conventional monoaminergic antidepressants are not fully effective. Therefore, ketamine has been accepted, with rigorous restrictions, in psychiatry as a new class of antidepressant. Notably, the dosage of ketamine for antidepressive action is comparable to the dose that can generate schizophrenia-like psychotic symptoms. Furthermore, the psychotropic effects of ketamine precede the antidepressant effects. The maintenance of the antidepressive efficacy of ketamine often requires repeated administration; however, repeated ketamine intake leads to abuse and is consistently associated with long-lasting memory-associated deficits. According to the dissociative anaesthetic feature of ketamine, it exerts broad acute influences on cognition/perception. To evaluate the therapeutic validation of ketamine across clinical contexts, including its advantages and disadvantages, psychiatry should systematically assess the safety and efficacy of either short- and long-term ketamine treatments, in terms of both acute and chronic outcomes. Here, we describe the clinical evidence of NMDAR antagonists, and then the temporal mechanisms of schizophrenia-like and antidepressant-like effects of the NMDAR antagonist, ketamine. The underlying pharmacological rodent studies will also be discussed.
Collapse
|
11
|
Adell A. Brain NMDA Receptors in Schizophrenia and Depression. Biomolecules 2020; 10:biom10060947. [PMID: 32585886 PMCID: PMC7355879 DOI: 10.3390/biom10060947] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 12/21/2022] Open
Abstract
N-methyl-D-aspartate (NMDA) receptor antagonists such as phencyclidine (PCP), dizocilpine (MK-801) and ketamine have long been considered a model of schizophrenia, both in animals and humans. However, ketamine has been recently approved for treatment-resistant depression, although with severe restrictions. Interestingly, the dosage in both conditions is similar, and positive symptoms of schizophrenia appear before antidepressant effects emerge. Here, we describe the temporal mechanisms implicated in schizophrenia-like and antidepressant-like effects of NMDA blockade in rats, and postulate that such effects may indicate that NMDA receptor antagonists induce similar mechanistic effects, and only the basal pre-drug state of the organism delimitates the overall outcome. Hence, blockade of NMDA receptors in depressive-like status can lead to amelioration or remission of symptoms, whereas healthy individuals develop psychotic symptoms and schizophrenia patients show an exacerbation of these symptoms after the administration of NMDA receptor antagonists.
Collapse
Affiliation(s)
- Albert Adell
- Institute of Biomedicine and Biotechnology of Cantabria, IBBTEC (CSIC-University of Cantabria), Calle Albert Einstein 22 (PCTCAN), 39011 Santander, Spain; or
- Biomedical Research Networking Center for Mental Health (CIBERSAM), 39011 Santander, Spain
| |
Collapse
|
12
|
Westlund KN, Lu Y, Zhang L, Pappas TC, Zhang WR, Taglialatela G, McIlwrath SL, McNearney TA. Tyrosine Kinase Inhibitors Reduce NMDA NR1 Subunit Expression, Nuclear Translocation, and Behavioral Pain Measures in Experimental Arthritis. Front Physiol 2020; 11:440. [PMID: 32536874 PMCID: PMC7267073 DOI: 10.3389/fphys.2020.00440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/08/2020] [Indexed: 11/17/2022] Open
Abstract
In the lumbar spinal cord dorsal horn, release of afferent nerve glutamate activates the neurons that relay information about injury pain. Here, we examined the effects of protein tyrosine kinase (PTK) inhibition on NMDA receptor NR1 subunit protein expression and subcellular localization in an acute experimental arthritis model. PTK inhibitors genistein and lavendustin A reduced cellular histological translocation of NMDA NR1 in the spinal cord occurring after the inflammatory insult and the nociceptive behavioral responses to heat. The PTK inhibitors were administered into lumbar spinal cord by microdialysis, and secondary heat hyperalgesia was determined using the Hargreaves test. NMDA NR1 cellular protein expression and nuclear translocation were determined by immunocytochemical localization with light and electron microscopy, as well as with Western blot analysis utilizing both C- and N-terminal antibodies. Genistein and lavendustin A (but not inactive lavendustin B or diadzein) effectively reduced (i) pain related behavior, (ii) NMDA NR1 subunit expression increases in spinal cord, and (iii) the shift of NR1 from a cell membrane to a nuclear localization. Genistein pre-treatment reduced these events that occur in vivo within 4 h after inflammatory insult to the knee joint with kaolin and carrageenan (k/c). Cycloheximide reduced glutamate activated upregulation of NR1 content confirming synthesis of new protein in response to the inflammatory insult. In addition to this in vivo data, genistein or staurosporin inhibited upregulation of NMDA NR1 protein and nuclear translocation in vitro after treatment of human neuroblastoma clonal cell cultures (SH-SY5Y) with glutamate or NMDA (4 h). These studies provide evidence that inflammatory activation of peripheral nerves initiates increase in NMDA NR1 in the spinal cord coincident with development of pain related behaviors through glutamate non-receptor, PTK dependent cascades.
Collapse
Affiliation(s)
- Karin N Westlund
- Research Division, New Mexico VA Health Care System, Albuquerque, NM, United States.,Anesthesiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States.,Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ying Lu
- Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Liping Zhang
- Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Todd C Pappas
- Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Wen-Ru Zhang
- Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Giulio Taglialatela
- Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States.,Neurology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Sabrina L McIlwrath
- Research Division, New Mexico VA Health Care System, Albuquerque, NM, United States
| | - Terry A McNearney
- Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States.,Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, United States.,Internal Medicine, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| |
Collapse
|
13
|
Quantitative analysis of Gria1, Gria2, Dlg1 and Dlg4 expression levels in hippocampus following forced swim stress in mice. Sci Rep 2019; 9:14060. [PMID: 31575955 PMCID: PMC6773768 DOI: 10.1038/s41598-019-50689-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/18/2019] [Indexed: 01/09/2023] Open
Abstract
AMPA receptors and interacting proteins are importantly involved in mediating stress-dependent plasticity. Previously we reported that GluA1-containing AMPA receptors and their interaction with PDZ-proteins are required for the experience-dependent expression of behavioral despair in the forced swim test. However, it is unclear if the expression of GluA1-containing AMPA receptors is affected by this type of behavior. Here we investigated in wild type mice, whether hippocampal gene or protein levels of GluA1 or associated PDZ proteins is altered following forced swim stress. We show that expression of Dlg4 (the gene coding for PSD-95) was strongly reduced after two days of forced swimming. In contrast, levels of Dlg1, Gria1, and Gria2 (coding for SAP97, GluA1, and GluA2 respectively) were not affected after one or two days of forced swimming. The changes in gene expression largely did not translate to the protein level. These findings indicate a limited acute effect of forced swim stress on the expression of the investigated targets and suggest that the acute involvement of GluA1-containing AMPA receptors tor forced swim behavior is a result of non-genomic mechanisms.
Collapse
|
14
|
Pérez-Valenzuela C, Terreros G, Dagnino-Subiabre A. Effects of stress on the auditory system: an approach to study a common origin for mood disorders and dementia. Rev Neurosci 2019; 30:317-324. [PMID: 30205652 DOI: 10.1515/revneuro-2018-0018] [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: 03/01/2018] [Accepted: 05/12/2018] [Indexed: 12/22/2022]
Abstract
The concept of stress is a fundamental piece to understand how organisms can adapt to the demands produced by a continuously changing environment. However, modern lifestyle subjects humans to high levels of negative stress or distress, which increases the prevalence of mental illnesses. Definitely, stress has become the pandemic of the 21st century, a fact that demands a great intellectual effort from scientists to understand the neurobiology of stress. This review proposes an innovative point of view to understand that mood disorders and dementia have a common etiology in a stressful environment. We propose that distress produces sensory deprivation, and this interferes with the connection between the brain and the environment in which the subject lives. The auditory system can serve as an example to understand this idea. In this sense, distress impairs the auditory system and induces hearing loss or presbycusis at an early age; this can increase the cognitive load in stressed people, which can stimulate the development of dementia in them. On the other hand, distress impairs the auditory system and increases the excitability of the amygdala, a limbic structure involved in the emotional processing of sounds. A consequence of these alterations could be the increase in the persistence of auditory fear memory, which could increase the development of mood disorders. Finally, it is important to emphasize that stress is an evolutionary issue that is necessary to understand the mental health of humans in these modern times. This article is a contribution to this discussion and will provide insights into the origin of stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Catherine Pérez-Valenzuela
- Laboratory of Stress Neurobiology, Institute of Physiology, Faculty of Sciences, Universidad de Valparaíso, Gran Bretaña 1111, Playa Ancha, Valparaíso, Chile
| | - Gonzalo Terreros
- Instituto de Ciencias de la Salud, Universidad de O'Higgins, Rancagua, Chile.,Auditory and Cognition Center (AUCO), Santiago, Chile
| | - Alexies Dagnino-Subiabre
- Laboratory of Stress Neurobiology, Institute of Physiology, Faculty of Sciences, Universidad de Valparaíso, Gran Bretaña 1111, Playa Ancha, Valparaíso, Chile.,Auditory and Cognition Center (AUCO), Santiago, Chile
| |
Collapse
|
15
|
Marrocco J, Gray JD, Kogan JF, Einhorn NR, O’Cinneide EM, Rubin TG, Carroll TS, Schmidt EF, McEwen BS. Early Life Stress Restricts Translational Reactivity in CA3 Neurons Associated With Altered Stress Responses in Adulthood. Front Behav Neurosci 2019; 13:157. [PMID: 31354448 PMCID: PMC6637287 DOI: 10.3389/fnbeh.2019.00157] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 06/27/2019] [Indexed: 12/27/2022] Open
Abstract
Early life experiences program brain structure and function and contribute to behavioral endophenotypes in adulthood. Epigenetic control of gene expression by those experiences affect discrete brain regions involved in mood, cognitive function and regulation of hypothalamic-pituitary-adrenal (HPA) axis. In rodents, acute restraint stress increases the expression of the repressive histone H3 lysine 9 tri-methylation (H3K9me3) in hippocampal fields, including the CA3 pyramidal neurons. These CA3 neurons are crucially involved in cognitive function and mood regulation as well as activation of glucocorticoid (CORT) secretion. CA3 neurons also exhibit structural and functional changes after early-life stress (ELS) as well as after chronic stress in adulthood. Using a protocol of chronic ELS induced by limited bedding and nesting material followed by acute-swim stress (AS) in adulthood, we show that mice with a history of ELS display a blunted CORT response to AS, despite exhibiting activation of immediate early genes after stress similar to that found in control mice. We find that ELS induced persistently increased expression of the repressive H3K9me3 histone mark in the CA3 subfield at baseline that was subsequently decreased following AS. In contrast, AS induced a transient increase of this mark in control mice. Using translating ribosome affinity purification (TRAP) method to isolate CA3 translating mRNAs, we found that expression of genes of the epigenetic gene family, GABA/glutamate family, and glucocorticoid receptors binding genes were decreased transiently in control mice by AS and showed a persistent reduction in ELS mice. In most cases, AS in ELS mice did not induce gene expression changes. A stringent filtering of genes affected by AS in control and ELS mice revealed a noteworthy decrease in gene expression change in ELS mice compared to control. Only 18 genes were selectively regulated by AS in ELS mice and encompassed pathways such as circadian rhythm, inflammatory response, opioid receptors, and more genes included in the glucocorticoid receptor binding family. Thus, ELS programs a restricted translational response to stress in stress-sensitive CA3 neurons leading to persistent changes in gene expression, some of which mimic the transient effects of AS in control mice, while leaving in operation the immediate early gene response to AS.
Collapse
Affiliation(s)
- Jordan Marrocco
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, United States
| | - Jason D. Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, United States
| | - Joshua F. Kogan
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, United States
| | - Nathan R. Einhorn
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, United States
| | - Emma M. O’Cinneide
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, United States
| | - Todd G. Rubin
- Albert Einstein College of Medicine, Bronx, NY, United States
| | - Thomas S. Carroll
- Bioinformatics Resource Center, The Rockefeller University, New York, NY, United States
| | - Eric F. Schmidt
- Laboratory of Molecular Biology, The Rockefeller University, New York, NY, United States
| | - Bruce S. McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, United States
| |
Collapse
|
16
|
Underlying mechanisms of recombinant adeno-associated virus-mediated bicaudal C homolog 1 overexpression in the medial prefrontal cortex of mice with induced depressive-like behaviors. Brain Res Bull 2019; 150:35-41. [PMID: 31102751 DOI: 10.1016/j.brainresbull.2019.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 11/22/2022]
Abstract
Bicaudal C homolog 1 gene (BICC1) in the medial prefrontal cortex (mPFC) has been implicated in major depressive disorder (MDD); however, less is known about the mechanisms of BICC1-induced depression. The purpose of the present study was to investigate changes in depressive-like behaviors induced by recombinant adeno-associated virus (rAAV)-mediated overexpression of BICC1 in the mPFC of mice. A viral-mediated genetic approach was employed to explore the BICC1 overexpression-induced depressive-like behavioral and molecular changes in mice. For the first time, we found that BICC1 overexpression significantly induced depressive-like behaviors in mice. Further, the expression of disheveled-2 and the phosphorylation of Ser9 of glycogen synthase kinase 3β (GSK3β), mechanistic target of rapamycin (mTOR) and GluA1, GluA1, brain-derived neurotrophic factor (BDNF), and VGF were markedly down-regulated in BICC1 overexpression-treated animals. Our results demonstrate that the overexpression of BICC1 in the mPFC may induce depressive-like behaviors via GSK3β/mTOR signaling and GluA1 trafficking in the mPFC of mice, indicating that BICC1 may be a potential target for antidepressant treatment.
Collapse
|
17
|
Wilkinson ST, Kiselycznyk C, Banasr M, Webler RD, Haile C, Mathew SJ. Serum and plasma brain-derived neurotrophic factor and response in a randomized controlled trial of riluzole for treatment resistant depression. J Affect Disord 2018; 241:514-518. [PMID: 30153634 PMCID: PMC6140345 DOI: 10.1016/j.jad.2018.08.075] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/02/2018] [Accepted: 08/12/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Serum brain-derived neurotrophic factor (BDNF) is decreased in individuals with major depressive disorder (MDD). Pre-clinical and clinical reports suggest that the glutamate release inhibitor riluzole increases BDNF and may have antidepressant properties. Here we report serum (sBDNF) and plasma (pBDNF) levels from a randomized controlled, adjunctive, sequential parallel comparison design trial of riluzole in MDD. METHODS Serum and plasma BDNF samples were drawn at baseline and weeks 6 and 8 from 55 subjects randomized to adjunctive treatment with riluzole or placebo for 8 weeks. RESULTS Riluzole responders had lower baseline serum (19.08 ng/ml [SD 9.22] v. 28.80 ng/ml [9.63], p = 0.08) and plasma (2.72 ng/ml [1.07] v. 4.60 ng/ml [1.69], p = 0.06) BDNF compared to non-responders at a trend level. This pattern was nominally seen in placebo responders for baseline pBDNF to some degree (1.21 ng/ml [SD 1.29] v. 3.58 ng/ml [SD 1.67], p = 0.12) but not in baseline sBDNF. LIMITATIONS A number of limitations warrant comment, including the small sample size of viable BDNF samples and the small number of riluzole responders. CONCLUSIONS Preliminary evidence reported here suggests that lower baseline BDNF may be associated with better clinical response to riluzole.
Collapse
Affiliation(s)
| | | | - Mounira Banasr
- Campbell Family Mental Health Research Institute of CAMH (Centre of Addiction and Mental Health), Toronto, ON, Canada
| | | | - Colin Haile
- Baylor College of Medicine, Houston, TX,Michael E. DeBakey VA Medical Center, Houston, TX
| | - Sanjay J. Mathew
- Baylor College of Medicine, Houston, TX,Michael E. DeBakey VA Medical Center, Houston, TX
| |
Collapse
|
18
|
Sunstrum JK, Inoue W. Heterosynaptic modulation in the paraventricular nucleus of the hypothalamus. Neuropharmacology 2018; 154:87-95. [PMID: 30408488 DOI: 10.1016/j.neuropharm.2018.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/18/2018] [Accepted: 11/03/2018] [Indexed: 12/21/2022]
Abstract
The stress response-originally described by Hans Selye as "the nonspecific response of the body to any demand made upon it"-is chiefly mediated by the hypothalamic-pituitary-adrenal (HPA) axis and is activated by diverse sensory stimuli that inform threats to homeostasis. The diversity of signals regulating the HPA axis is partly achieved by the complexity of afferent inputs that converge at the apex of the HPA axis: this apex is formed by a group of neurosecretory neurons that synthesize corticotropin-releasing hormone (CRH) in the paraventricular nucleus of the hypothalamus (PVN). The afferent synaptic inputs onto these PVN-CRH neurons originate from a number of brain areas, and PVN-CRH neurons respond to a long list of neurotransmitters/neuropeptides. Considering this complexity, an important question is how these diverse afferent signals independently and/or in concert influence the excitability of PVN-CRH neurons. While many of these inputs directly act on the postsynaptic PVN-CRH neurons for the summation of signals, accumulating data indicates that they also modulate each other's transmission in the PVN. This mode of transmission, termed heterosynaptic modulation, points to mechanisms through which the activity of a specific modulatory input (conveying a specific sensory signal) can up- or down-regulate the efficacy of other afferent synapses (mediating other stress modalities) depending on receptor expression for and spatial proximity to the heterosynaptic signals. Here, we review examples of heterosynaptic modulation in the PVN and discuss its potential role in the regulation of PVN-CRH neurons' excitability and resulting HPA axis activity. This article is part of the Special Issue entitled 'Hypothalamic Control of Homeostasis'.
Collapse
Affiliation(s)
- Julia K Sunstrum
- Neuroscience Program, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Wataru Inoue
- Neuroscience Program, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
19
|
Lan Z, Xu J, Wang Y, Lu W. Modulatory effect of glutamate GluR2 receptor on the caudal neurosecretory Dahlgren cells of the olive flounder, Paralichthys olivaceus. Gen Comp Endocrinol 2018; 261:9-22. [PMID: 29355533 DOI: 10.1016/j.ygcen.2018.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/13/2018] [Accepted: 01/14/2018] [Indexed: 11/26/2022]
Abstract
A neuromodulatory role for glutamate has been reported for magnocellular neuroendocrine cells in mammalian hypothalamus. We examined the potential role of glutamate as a local intercellular messenger in the neuroendocrine Dahlgren cell population of the caudal neurosecretory system (CNSS) in the euryhaline flounder Paralichthys olivaceus. In pharmacological experiments in vitro, glutamate (Glu) caused an increase in electrical activity of Dahlgren cells, recruitment of previously silent cells, together with a greater proportion of cells showing phasic (irregular) activity. The glutamate substrate, glutamine (Gln), led to increased firing frequency, cell recruitment and enhanced bursting activity. The glutamate effect was not blocked by the N-methyl-D-aspartate (NMDA) receptor antagonist MK-801, or the GluR1/GluR3 (AMPA) receptor antagonist IEm1795-2HBr, but was blocked by the broad-spectrum α-amino-3-hydroxy- 5- methyl-4-isoxazo-lepropionic acid (AMPA) receptor antagonist ZK200775. Our transcriptome sequencing study revealed three AMPA receptor (GluR1, GluR2 and GluR3) in the olive flounder CNSS. Quantitative RT-PCR revealed that GluR2 receptor mRNA expression was significant increased following dose-dependent superfusion with glutamate in the CNSS. GluR1 and GluR3 receptor mRNA expression were decreased following superfusion with glutamate. L-type Ca2+ channel mRNA expression had a significant dose-dependent decrease following superfusion with glutamate, compared to the control. In the salinity challenge experiment, acute transfer from SW to FW, GluR2 receptor mRNA expression was significantly higher than the control at 2 h. These findings suggest that GluR2 is one of the mechanisms which can medicate glutamate action within the CNSS, enhancing electrical activity and hence secretory output.
Collapse
Affiliation(s)
- Zhaohui Lan
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Jinling Xu
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Youji Wang
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, China
| | - Weiqun Lu
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, China.
| |
Collapse
|
20
|
Translational profiling of stress-induced neuroplasticity in the CA3 pyramidal neurons of BDNF Val66Met mice. Mol Psychiatry 2018; 23:904-913. [PMID: 27956743 PMCID: PMC5468507 DOI: 10.1038/mp.2016.219] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/16/2016] [Accepted: 10/14/2016] [Indexed: 02/07/2023]
Abstract
Genetic susceptibility and environmental factors (such as stress) can interact to affect the likelihood of developing a mood disorder. Stress-induced changes in the hippocampus have been implicated in mood disorders, and mutations in several genes have now been associated with increased risk, such as brain-derived neurotrophic factor (BDNF). The hippocampus has important anatomical subdivisions, and pyramidal neurons of the vulnerable CA3 region show significant remodeling after chronic stress, but the mechanisms underlying their unique plasticity remain unknown. This study characterizes stress-induced changes in the in vivo translating mRNA of this cell population using a CA3-specific enhanced green fluorescent protein (EGFP) reporter fused to the L10a large ribosomal subunit (EGFPL10a). RNA-sequencing after isolation of polysome-bound mRNAs allows for cell-type-specific, genome-wide characterization of translational changes after stress. The data demonstrate that acute and chronic stress produce unique translational profiles and that the stress history of the animal can alter future reactivity of CA3 neurons. CA3-specific EGFPL10a mice were then crossed to the stress-susceptible BDNF Val66Met mouse line to characterize how a known genetic susceptibility alters both baseline translational profiles and the reactivity of CA3 neurons to stress. Not only do Met allele carriers exhibit distinct levels of baseline translation in genes implicated in ion channel function and cytoskeletal regulation, but they also activate a stress response profile that is highly dissimilar from wild-type mice. Closer examination of genes implicated in the mechanisms of neuroplasticity, such as the NMDA and AMPA subunits and the BDNF pathway, reveal how wild-type mice upregulate many of these genes in response to stress, but Met allele carriers fail to do so. These profiles provide a roadmap of stress-induced changes in a genetically homogenous population of hippocampal neurons and illustrate the profound effects of gene-environment interactions on the translational profile of these cells.
Collapse
|
21
|
Yasmin F, Saxena K, McEwen BS, Chattarji S. The delayed strengthening of synaptic connectivity in the amygdala depends on NMDA receptor activation during acute stress. Physiol Rep 2017; 4:4/20/e13002. [PMID: 27798355 PMCID: PMC5099964 DOI: 10.14814/phy2.13002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/20/2016] [Indexed: 01/21/2023] Open
Abstract
There is growing evidence that stress leads to contrasting patterns of structural plasticity in the hippocampus and amygdala, two brain areas implicated in the cognitive and affective symptoms of stress‐related psychiatric disorders. Acute stress has been shown to trigger a delayed increase in the density of dendritic spines in the basolateral amygdala (BLA) of rodents. However, the physiological correlates of this delayed spinogenesis in the BLA remain unexplored. Furthermore, NMDA receptors (NMDARs) have been known to underlie chronic stress‐induced structural plasticity in the hippocampus, but nothing is known about the role of these receptors in the delayed spinogenesis, and its physiological consequences, in the BLA following acute stress. Here, using whole‐cell recordings in rat brain slices, we find that a single exposure to 2‐h immobilization stress enhances the frequency, but not amplitude, of miniature excitatory postsynaptic currents (mEPSCs) recorded from principal neurons in the BLA 10 days later. This was also accompanied by faster use‐dependent block of NMDA receptor currents during repeated stimulation of thalamic inputs to the BLA, which is indicative of higher presynaptic release probability at these inputs 10 days later. Furthermore, targeted in vivo infusion of the NMDAR‐antagonist APV into the BLA during the acute stress prevents the increase in mEPSC frequency and spine density 10 days later. Together, these results identify a role for NMDARs during acute stress in both the physiological and morphological strengthening of synaptic connectivity in the BLA in a delayed fashion. These findings also raise the possibility that activation of NMDA receptors during stress may serve as a common molecular mechanism despite the divergent patterns of plasticity that eventually emerge after stress in the amygdala and hippocampus.
Collapse
Affiliation(s)
- Farhana Yasmin
- National Centre for Biological Sciences, Bangalore, India
| | - Kapil Saxena
- National Centre for Biological Sciences, Bangalore, India.,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, USA
| | - Sumantra Chattarji
- National Centre for Biological Sciences, Bangalore, India .,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.,Centre for Integrative Physiology, Deanery of Biomedical Sciences, The University of Edinburgh, George Square, Edinburgh, UK
| |
Collapse
|
22
|
Chronic-Stress-Induced Behavioral Changes Associated with Subregion-Selective Serotonin Cell Death in the Dorsal Raphe. J Neurosci 2017; 37:6214-6223. [PMID: 28546314 DOI: 10.1523/jneurosci.3781-16.2017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 04/25/2017] [Accepted: 05/16/2017] [Indexed: 01/18/2023] Open
Abstract
The current study examined the neurochemical mechanisms and neuroanatomical changes underlying coexisting behavioral effects associated with chronic-stress-induced alterations in serotonin (5HT) neurons. Chronic unpredictable stress (CUS) to adult male rats produced depression-like changes with cognitive dysfunction and selective cell death in the interfascicular nucleus of the dorsal raphe (DRif), resulting in decreased 5HTergic innervation of medial prefrontal cortex (mPFC). Twenty-one days of CUS decreased basal plasma levels of corticosterone and produced a shorter latency to immobility and longer durations of immobility in the force-swim test that persisted for 1 month after CUS. Deficits in acquisition, recall, perseveration, and reversal learning were evident 1 month after CUS. MK801 treatment during CUS blocked the changes in the forced-swim test and deficits in memory recall. These behavioral changes were associated with terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive soma and the eventual loss of 5HT neurons in the DRif and its projections to the mPFC as evidenced by fewer labeled cells in the DRif after retrograde tracer injections into the mPFC of stressed rats. Similar to the effects of MK801 on behavior, MK801 pretreatment during stress blocked the CUS-induced decreases in 5HT soma within the DRif and its projections to the mPFC. Finally, the depression-like behaviors were blocked by acute injection of the 5HT2A/C agonist (-)-2,5-dimethoxy-4-iodoamphetamine hydrochloride into the mPFC before forced-swim testing. These results identify a cause and mechanism of 5HTergic dysfunction of the mPFC and associated mood and cognitive behaviors.SIGNIFICANCE STATEMENT Chronic stress causes persistent mood and cognitive changes typically associated with dysregulated serotonin (5HT) transmission in the medial prefrontal cortex (mPFC), but the cause of this dysregulation is unknown. Prior studies have focused on 5HTergic terminals in this region, but this study shows that chronic stress causes NMDA-receptor-dependent and subregion-specific cell death of 5HT neurons in the dorsal raphe. The consequent decreased 5HT innervation of the mPFC was associated with mood and cognitive changes that persisted long after the termination of stress. These findings identify a mechanism of subregion-selective death of 5HT neurons in the dorsal raphe, a defined neuroanatomical pathway, and a behavioral phenotype that mirror stress-associated diseases such as major depressive disorder.
Collapse
|
23
|
Kusek M, Tokarska A, Siwiec M, Gadek-Michalska A, Szewczyk B, Hess G, Tokarski K. Nitric Oxide Synthase Inhibitor Attenuates the Effects of Repeated Restraint Stress on Synaptic Transmission in the Paraventricular Nucleus of the Rat Hypothalamus. Front Cell Neurosci 2017; 11:127. [PMID: 28515682 PMCID: PMC5413825 DOI: 10.3389/fncel.2017.00127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/18/2017] [Indexed: 11/29/2022] Open
Abstract
Corticotropin-releasing hormone (CRH)-synthesizing parvocellular neuroendocrine cells (PNCs) of the hypothalamic paraventricular nucleus (PVN) play a key role in the activation of the hypothalamic-pituitary-adrenocortical (HPA) axis. Several studies have demonstrated that synaptic inputs to these cells may undergo stress-related enhancement but, on the other hand, it has been reported that exposition to the same stressor for prolonged time periods may induce a progressive reduction in the response of the HPA axis to homotypic stressors. In the present study rats were subjected to 10 min restraint sessions, repeated twice daily for 3 or 7 days. Miniature excitatory and inhibitory postsynaptic currents (mEPSCs and mIPSCs) were then recorded from PNCs in ex vivo hypothalamic slice preparations obtained 24 h after the last restraint. Restraint stress repeated over 3 days resulted in increased mean frequency and decreased rise time and decay time constant of mEPSCs, accompanied by a decrease in the excitability of PNCs, however, no such changes were evident in slices obtained from rats subjected to restraint over 7 days. There were no changes in mIPSCs after repeated restraint. Administration of the unspecific nitric oxide synthase (NOS) blocker Nω-Nitro-L-arginine (L-NNA) before each restraint, repeated over 3 days, prevented the occurrence of an increase in mEPSC frequency. However, animals receiving L-NNA and subjected to repeated restraint had similar changes in PNCs membrane excitability and mEPSC kinetics as stressed rats not receiving L-NNA. Comparison of the effects of a single 10 min restraint session followed by either an immediate or delayed (24 h) decapitation revealed an increase in the mean mEPSC frequency and a decrease in the mean mIPSC frequency in slices prepared immediately after restraint, with no apparent effects when slice preparation was delayed by 24 h. These results demonstrate that restraint, lasting 10 min and repeated twice daily for 3 days, induces a selective and long-lasting enhancement of excitatory synaptic input onto PNCs, partially by a NOS-dependent mechanism, and reduces PNC excitability, whereas prolongation of repeated stress for up to 7 days results in an adaptation.
Collapse
Affiliation(s)
- Magdalena Kusek
- Department of Physiology, Institute of Pharmacology, Polish Academy of SciencesKraków, Poland
| | - Anna Tokarska
- Institute of Zoology and Biomedical Research, Jagiellonian UniversityKraków, Poland
| | - Marcin Siwiec
- Department of Physiology, Institute of Pharmacology, Polish Academy of SciencesKraków, Poland
| | - Anna Gadek-Michalska
- Department of Physiology, Institute of Pharmacology, Polish Academy of SciencesKraków, Poland
| | - Bernadeta Szewczyk
- Department of Neurobiology, Institute of Pharmacology, Polish Academy of SciencesKraków, Poland
| | - Grzegorz Hess
- Department of Physiology, Institute of Pharmacology, Polish Academy of SciencesKraków, Poland.,Institute of Zoology and Biomedical Research, Jagiellonian UniversityKraków, Poland
| | - Krzysztof Tokarski
- Department of Physiology, Institute of Pharmacology, Polish Academy of SciencesKraków, Poland
| |
Collapse
|
24
|
Mikics E, Toth M, Biro L, Bruzsik B, Nagy B, Haller J. The role of GluN2B-containing NMDA receptors in short- and long-term fear recall. Physiol Behav 2017; 177:44-48. [PMID: 28400283 DOI: 10.1016/j.physbeh.2017.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/24/2017] [Accepted: 04/07/2017] [Indexed: 12/27/2022]
Abstract
N-methyl-d-aspartate (NMDA) receptors are crucial synaptic elements in long-term memory formation, including the associative learning of fearful events. Although NMDA blockers were consistently shown to inhibit fear memory acquisition and recall, the clinical use of general NMDA blockers is hampered by their side effects. Recent studies revealed significant heterogeneity in the distribution and neurophysiological characteristics of NMDA receptors with different GluN2 (NR2) subunit composition, which may have differential role in fear learning and recall. To investigate the specific role of NMDA receptor subpopulations with different GluN2 subunit compositions in the formation of lasting traumatic memories, we contrasted the effects of general NMDA receptor blockade with GluN2A-, GluN2B-, and GluN2C/D subunit selective antagonists (MK-801, PEAQX, Ro25-6981, PPDA, respectively). To investigate acute and lasting consequences, behavioral responses were investigated 1 and 28days after fear conditioning. We found that MK-801 (0.05 and 0.1mg/kg) decreased fear recall at both time points. GluN2B receptor subunit blockade produced highly similar effects, albeit efficacy was somewhat smaller 28days after fear conditioning. Unlike MK-801, Ro25-6981 (3 and 10mg/kg) did not affect locomotor activity in the open-field. In contrast, GluN2A and GluN2C/D blockers (6 and 20mg/kg PEAQX; 3 and 10mg/kg PPDA, respectively) had no effect on conditioned fear recall at any time point and dose. This sharp contrast between GluN2B- and other subunit-containing NMDA receptor function indicates that GluN2B receptor subunits are intimately involved in fear memory formation, and may provide a novel pharmacological target in post-traumatic stress disorder or other fear-related disorders.
Collapse
Affiliation(s)
- Eva Mikics
- Institute of Experimental Medicine, Department of Behavioral Neuroscience, P.O. Box 67, H-1450 Budapest, Hungary.
| | - Mate Toth
- Institute of Experimental Medicine, Department of Behavioral Neuroscience, P.O. Box 67, H-1450 Budapest, Hungary
| | - Laszlo Biro
- Institute of Experimental Medicine, Department of Behavioral Neuroscience, P.O. Box 67, H-1450 Budapest, Hungary
| | - Biborka Bruzsik
- Institute of Experimental Medicine, Department of Behavioral Neuroscience, P.O. Box 67, H-1450 Budapest, Hungary
| | - Boglarka Nagy
- Institute of Experimental Medicine, Department of Behavioral Neuroscience, P.O. Box 67, H-1450 Budapest, Hungary
| | - Jozsef Haller
- Institute of Experimental Medicine, Department of Behavioral Neuroscience, P.O. Box 67, H-1450 Budapest, Hungary
| |
Collapse
|
25
|
Effects of (-)-sesamin on chronic stress-induced memory deficits in mice. Neurosci Lett 2016; 634:114-118. [DOI: 10.1016/j.neulet.2016.09.055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/08/2016] [Accepted: 09/29/2016] [Indexed: 01/14/2023]
|
26
|
Fokos S, Pavlidis M, Yiotis T, Tsalafouta A, Papandroulakis N, Dermon CR. Early life low intensity stress experience modifies acute stress effects on juvenile brain cell proliferation of European sea bass (D. Labrax). Behav Brain Res 2016; 317:109-121. [PMID: 27638037 DOI: 10.1016/j.bbr.2016.09.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 09/04/2016] [Accepted: 09/11/2016] [Indexed: 02/06/2023]
Abstract
Early life adversity may be critical for the brain structural plasticity that in turn would influence juvenile behaviour. To address this, we questioned whether early life environment has an impact on stress responses latter in life, using European sea bass, Dicentrarchus labrax, as a model organism. Unpredictable chronic low intensity stress (UCLIS), using a variety of moderate intensity stressors, was applied during two early ontogenetic stages, flexion or formation all fins. At juvenile stage, fish were exposed to acute stress and plasma cortisol, brain mRNA expression of corticosteroid receptors' genes (gr1, gr2, mr) and brain cell proliferation (using BrdU immunohistochemistry) were determined in experimental and matched controls. UCLIS treatment specifically decreased brain gr1 expression in juveniles, but had no effect on the juvenile brain cell proliferation pattern within the major neurogenic zones studied of dorsal (Dm, Dld) and ventral (Vv) telencephalic, preoptic (NPO) areas, periventricular tectum gray zone (PGZ) and valvula cerebellum (VCe). In contrast, exposure to acute stress induced significant plasma cortisol rise, decreases of cerebral cell proliferation in juveniles, not previously exposed to UCLIS, but no effect detected on the expression levels of gr1, gr2 and mr in all groups of different early life history. Interestingly, juveniles with UCLIS history showed modified responses to acute stress, attenuating acute stress-induced cell proliferation decreases, indicating a long-lasting effect of early life treatment. Taken together, early life mild stress experience influences an acute stress plasticity end-point, cerebral cell proliferation, independently of the stress-axis activation, possibly leading to more effective coping styles.
Collapse
Affiliation(s)
- S Fokos
- Dept. of Biology, Human and Animal Physiology Lab, University of Patras, Greece(1)
| | - M Pavlidis
- Dept. of Biology, University of Crete, Greece
| | - T Yiotis
- Dept. of Biology, Human and Animal Physiology Lab, University of Patras, Greece(1)
| | - A Tsalafouta
- Dept. of Biology, University of Crete, Greece; Aquaculture Institute, Hellenic Centre Marine Research, Crete, Greece
| | - N Papandroulakis
- Aquaculture Institute, Hellenic Centre Marine Research, Crete, Greece
| | - C R Dermon
- Dept. of Biology, Human and Animal Physiology Lab, University of Patras, Greece(1).
| |
Collapse
|
27
|
Novick AM, Mears M, Forster GL, Lei Y, Tejani-Butt SM, Watt MJ. Adolescent social defeat alters N-methyl-D-aspartic acid receptor expression and impairs fear learning in adulthood. Behav Brain Res 2016; 304:51-9. [PMID: 26876136 PMCID: PMC4795455 DOI: 10.1016/j.bbr.2016.02.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 01/27/2016] [Accepted: 02/08/2016] [Indexed: 11/19/2022]
Abstract
Repeated social defeat of adolescent male rats results in adult mesocortical dopamine hypofunction, impaired working memory, and increased contextual anxiety-like behavior. Given the role of glutamate in dopamine regulation, cognition, and fear and anxiety, we investigated potential changes to N-methyl-D-aspartic acid (NMDA) receptors following adolescent social defeat. As both NMDA receptors and mesocortical dopamine are implicated in the expression and extinction of conditioned fear, a separate cohort of rats was challenged with a classical fear conditioning paradigm to investigate whether fear learning is altered by adolescent defeat. Quantitative autoradiography was used to measure 3H-MK-801 binding to NMDA receptors in regions of the medial prefrontal cortex, caudate putamen, nucleus accumbens, amygdala and hippocampus. Assessment of fear learning was achieved using an auditory fear conditioning paradigm, with freezing toward the auditory tone used as a measure of conditioned fear. Compared to controls, adolescent social defeat decreased adult NMDA receptor expression in the infralimbic region of the prefrontal cortex and central amygdala, while increasing expression in the CA3 region of the hippocampus. Previously defeated rats also displayed decreased conditioned freezing during the recall and first extinction periods, which may be related to the observed decreases and increases in NMDA receptors within the central amygdala and CA3, respectively. The alteration in NMDA receptors seen following adolescent social defeat suggests that dysfunction of glutamatergic systems, combined with mesocortical dopamine deficits, likely plays a role in the some of the long-term behavioral consequences of social stressors in adolescence seen in both preclinical and clinical studies.
Collapse
Affiliation(s)
- Andrew M Novick
- Center for Brain and Behavior Research, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, 414 E. Clark St., Vermillion, SD 57069, USA.
| | - Mackenzie Mears
- Center for Brain and Behavior Research, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, 414 E. Clark St., Vermillion, SD 57069, USA
| | - Gina L Forster
- Center for Brain and Behavior Research, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, 414 E. Clark St., Vermillion, SD 57069, USA
| | - Yanlin Lei
- Department of Pharmaceutical Sciences, University of the Sciences in Philadelphia, 600 S 43rd St., Philadelphia, PA 19104, USA
| | - Shanaz M Tejani-Butt
- Department of Pharmaceutical Sciences, University of the Sciences in Philadelphia, 600 S 43rd St., Philadelphia, PA 19104, USA
| | - Michael J Watt
- Center for Brain and Behavior Research, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, 414 E. Clark St., Vermillion, SD 57069, USA
| |
Collapse
|
28
|
|
29
|
Wang Y, Ma Y, Hu J, Cheng W, Jiang H, Zhang X, Li M, Ren J, Li X. Prenatal chronic mild stress induces depression-like behavior and sex-specific changes in regional glutamate receptor expression patterns in adult rats. Neuroscience 2015; 301:363-74. [DOI: 10.1016/j.neuroscience.2015.06.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 06/03/2015] [Accepted: 06/04/2015] [Indexed: 12/31/2022]
|
30
|
Kaut O, Schmitt I, Hofmann A, Hoffmann P, Schlaepfer TE, Wüllner U, Hurlemann R. Aberrant NMDA receptor DNA methylation detected by epigenome-wide analysis of hippocampus and prefrontal cortex in major depression. Eur Arch Psychiatry Clin Neurosci 2015; 265:331-41. [PMID: 25571874 DOI: 10.1007/s00406-014-0572-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 12/19/2014] [Indexed: 12/21/2022]
Abstract
Current perspectives on the molecular underpinnings of major depressive disorder (MDD) posit a mechanistic role of epigenetic DNA modifications in mediating the interaction between environmental risk factors and a genetic predisposition. However, conclusive evidence for differential methylation signatures in the brain's epigenome of MDD patients as compared to controls is still lacking. To address this issue, we conducted a pilot study including an epigenome-wide methylation analysis in six individuals diagnosed with recurrent MDD and six control subjects matched for age and gender, with a priori focus on the hippocampus and prefrontal cortex as pathophysiologically relevant candidate regions. Our analysis revealed differential methylation profiles of 11 genes in hippocampus and 20 genes in prefrontal cortex, five of which were selected for replication of the methylation status using pyrosequencing. Among these replicated targets, GRIN2A was found to be hypermethylated in both prefrontal cortex and hippocampus. This finding may be of particular functional relevance as GRIN2A encodes the glutamatergic N-methyl-D-aspartate receptor subunit epsilon-1 (NR2A) and is known to be involved in a plethora of synaptic plasticity-related regulatory processes probably disturbed in MDD.
Collapse
Affiliation(s)
- Oliver Kaut
- Department of Neurology, University of Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany,
| | | | | | | | | | | | | |
Collapse
|
31
|
Mayhew J, Beart PM, Walker FR. Astrocyte and microglial control of glutamatergic signalling: a primer on understanding the disruptive role of chronic stress. J Neuroendocrinol 2015; 27:498-506. [PMID: 25737228 DOI: 10.1111/jne.12273] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 02/23/2015] [Accepted: 02/27/2015] [Indexed: 01/23/2023]
Abstract
It is now well established that chronic stress can induce significant structural remodelling of astrocytes and microglia. Until recently, however, the full significance of these morphological disturbances has remained unclear. Clues to the significance of astroglial re-organisation following stress are beginning to emerge from a compelling literature describing how astrocytes contribute to glutamatergic neurotransmission. The present review briefly summarises these two fields of research, identifies points of overlap and, in doing so, pin-points future research directions for stress neurobiology. Ultimately, understanding how chronic stress can disrupt the interactions of astrocytes and microglia with neurones has the potential in the future to improve the development of therapeutics designed to treat stress-related illnesses such as depression.
Collapse
Affiliation(s)
- J Mayhew
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Centre for Translational Neuroscience and Mental Health Research, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - P M Beart
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Vic., Australia
| | - F R Walker
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Centre for Translational Neuroscience and Mental Health Research, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
32
|
Thompson SM, Kallarackal AJ, Kvarta MD, Van Dyke AM, LeGates TA, Cai X. An excitatory synapse hypothesis of depression. Trends Neurosci 2015; 38:279-94. [PMID: 25887240 DOI: 10.1016/j.tins.2015.03.003] [Citation(s) in RCA: 214] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 02/23/2015] [Accepted: 03/17/2015] [Indexed: 12/14/2022]
Abstract
Depression is a common cause of mortality and morbidity, but the biological bases of the deficits in emotional and cognitive processing remain incompletely understood. Current antidepressant therapies are effective in only some patients and act slowly. Here, we propose an excitatory synapse hypothesis of depression in which chronic stress and genetic susceptibility cause changes in the strength of subsets of glutamatergic synapses at multiple locations, including the prefrontal cortex (PFC), hippocampus, and nucleus accumbens (NAc), leading to a dysfunction of corticomesolimbic reward circuitry that underlies many of the symptoms of depression. This hypothesis accounts for current depression treatments and suggests an updated framework for the development of better therapeutic compounds.
Collapse
Affiliation(s)
- Scott M Thompson
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA; Department of Psychiatry, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA; Programs in Neuroscience and Membrane Biology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA.
| | - Angy J Kallarackal
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA; Programs in Neuroscience and Membrane Biology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA
| | - Mark D Kvarta
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA; Programs in Neuroscience and Membrane Biology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA; Medical Scientist Training Program, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA
| | - Adam M Van Dyke
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA; Programs in Neuroscience and Membrane Biology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA
| | - Tara A LeGates
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA
| | - Xiang Cai
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA; Department of Physiology, Southern Illinois University, Carbondale, IL 62901, USA
| |
Collapse
|
33
|
Freudenberg F, Celikel T, Reif A. The role of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in depression: central mediators of pathophysiology and antidepressant activity? Neurosci Biobehav Rev 2015; 52:193-206. [PMID: 25783220 DOI: 10.1016/j.neubiorev.2015.03.005] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 02/23/2015] [Accepted: 03/06/2015] [Indexed: 12/27/2022]
Abstract
Depression is a major psychiatric disorder affecting more than 120 million people worldwide every year. Changes in monoaminergic transmitter release are suggested to take part in the pathophysiology of depression. However, more recent experimental evidence suggests that glutamatergic mechanisms might play a more central role in the development of this disorder. The importance of the glutamatergic system in depression was particularly highlighted by the discovery that N-methyl-D-aspartate (NMDA) receptor antagonists (particularly ketamine) exert relatively long-lasting antidepressant like effects with rapid onset. Importantly, the antidepressant-like effects of NMDA receptor antagonists, but also other antidepressants (both classical and novel), require activation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Additionally, expression of AMPA receptors is altered in patients with depression. Moreover, preclinical evidence supports an important involvement of AMPA receptor-dependent signaling and plasticity in the pathophysiology and treatment of depression. Here we summarize work published on the involvement of AMPA receptors in depression and discuss a possible central role for AMPA receptors in the pathophysiology, course and treatment of depression.
Collapse
Affiliation(s)
- Florian Freudenberg
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Frankfurt, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany.
| | - Tansu Celikel
- Department of Neurophysiology, Donders Center for Neuroscience, Radboud University Nijmegen, 6500 AA Nijmegen, The Netherlands
| | - Andreas Reif
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Frankfurt, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany
| |
Collapse
|
34
|
Guercio GD, Bevictori L, Vargas-Lopes C, Madeira C, Oliveira A, Carvalho VF, d'Avila JC, Panizzutti R. D-serine prevents cognitive deficits induced by acute stress. Neuropharmacology 2014; 86:1-8. [PMID: 24978104 DOI: 10.1016/j.neuropharm.2014.06.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 06/11/2014] [Accepted: 06/19/2014] [Indexed: 01/03/2023]
Abstract
Increasing evidence indicates that acute stress disrupts cognitive functions mediated by glutamate-NMDA receptors, although the mechanisms are not fully understood. Here we investigated whether d-serine and glycine, the endogenous co-agonists of the NMDA receptor, are regulated by acute stress. We studied the biochemical and behavioral effects of acute restraint stress in C57BL/6 mice. Acute restraint stress decreased d-serine levels in the prefrontal cortex and glycine levels in the hippocampus. Behaviorally, acute stress impaired memory consolidation in the object recognition task and prepulse inhibition of the startle response. Importantly, d-serine administration (1 g/kg, i.p.) prevented both stress-induced impairments. Taken together, our results show for the first time an interplay between stress and d-serine and warrant further research on the role of d-serine in stress-related disorders.
Collapse
Affiliation(s)
- G D Guercio
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Brazil
| | - L Bevictori
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Brazil
| | - C Vargas-Lopes
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Brazil
| | - C Madeira
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Brazil
| | - A Oliveira
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Brazil
| | - V F Carvalho
- Oswaldo Cruz Foundation, FIOCRUZ, Rio de Janeiro, Brazil
| | - J C d'Avila
- Oswaldo Cruz Foundation, FIOCRUZ, Rio de Janeiro, Brazil
| | - R Panizzutti
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Brazil.
| |
Collapse
|
35
|
Effects of CA1 glutamatergic systems upon memory impairments in cholestatic rats. Behav Brain Res 2013; 256:636-45. [PMID: 24050889 DOI: 10.1016/j.bbr.2013.08.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 08/05/2013] [Accepted: 08/09/2013] [Indexed: 12/30/2022]
Abstract
BACKGROUND Bile duct ligation (BDL) is shown to induce cholestasis-related liver function impairments as well as consequent cognitive dysfunctions (i.e. impaired learning and memory formation). Glutamatergic neurotransmission plays an important role in hippocampal modulation of learning and memory function. The present study aimed to investigate the possible involvement of dorsal hippocampal (CA1) glutamatergic systems upon cholestasis-induced amnesia. METHOD Cholestasis was induced in male Wistar rats through double-ligation of the main bile duct (at two points) and transection of the interposed segment. Step-through passive avoidance test was employed to examine rats' learning and memory function. All drugs were injected into CA1 region of the hippocampus. RESULTS our results indicated a decrease in memory retrieval following cholestasis (11, 17 and 24 days post BDL). Only subthreshold doses of N-methyl-d-aspartate (NMDA; 0.125 and 0.25 μg/μl) but not its effective dose (0.5 μg/μl), restored the cholestasis-induced amnesia in step-through passive avoidance test, 11, 17 and 24 days post BDL. This effect was blocked by the subthreshold dose of D-[1]-2-amino-7-phosphonoheptanoic acid (D-AP7, NMDA receptor antagonist; 0.0625 μg/μl, intra-CA1) at 0.125 μg/μl and 0.25 μg/μl doses of NMDA. Moreover, our data revealed that only effective doses of D-AP7 (0.125 and 0.25 μg/μl, intra-CA1) potentiate memory impairments in 11 days after BDL. It was noted that none of applied drugs/doses exerted an effect on memory acquisition and locomotors activity, 10 and 12 days post laparotomy, respectively. CONCLUSION Our findings suggest the potential involvement of CA1 glutamatergic system(s) in cholestasis-induced memory deficits.
Collapse
|
36
|
Daskalakis NP, Yehuda R, Diamond DM. Animal models in translational studies of PTSD. Psychoneuroendocrinology 2013; 38:1895-911. [PMID: 23845512 DOI: 10.1016/j.psyneuen.2013.06.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 06/05/2013] [Accepted: 06/05/2013] [Indexed: 01/29/2023]
Abstract
Understanding the neurobiological mechanisms of post-traumatic stress disorder (PTSD) is of vital importance for developing biomarkers and more effective pharmacotherapy for this disorder. The design of bidirectional translational studies addressing all facets of PTSD is needed. Animal models of PTSD are needed not only to capture the complexity of PTSD behavioral characteristics, but also to address experimentally the influence of variety of factors which might determine an individual's vulnerability or resilience to trauma, e.g., genetic predisposition, early-life experience and social support. The current review covers recent translational approaches to bridge the gap between human and animal PTSD research and to create a framework for discovery of biomarkers and novel therapeutics.
Collapse
Affiliation(s)
- Nikolaos P Daskalakis
- Traumatic Stress Studies Division & Laboratory of Molecular Neuropsychiatry, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA; Mental Health Care Center, PTSD Clinical Research Program & Laboratory of Clinical Neuroendocrinology and Neurochemistry, James J. Peters Veterans Affairs Medical Center, Bronx, USA
| | | | | |
Collapse
|
37
|
Zoladz PR, Fleshner M, Diamond DM. Differential effectiveness of tianeptine, clonidine and amitriptyline in blocking traumatic memory expression, anxiety and hypertension in an animal model of PTSD. Prog Neuropsychopharmacol Biol Psychiatry 2013; 44:1-16. [PMID: 23318688 DOI: 10.1016/j.pnpbp.2013.01.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 12/22/2012] [Accepted: 01/04/2013] [Indexed: 12/21/2022]
Abstract
Individuals exposed to life-threatening trauma are at risk for developing post-traumatic stress disorder (PTSD), a debilitating condition that involves persistent anxiety, intrusive memories and several physiological disturbances. Current pharmacotherapies for PTSD manage only a subset of these symptoms and typically have adverse side effects which limit their overall effectiveness. We evaluated the effectiveness of three different pharmacological agents to ameliorate a broad range of PTSD-like symptoms in our established predator-based animal model of PTSD. Adult male Sprague-Dawley rats were given 1-h cat exposures on two occasions that were separated by 10 days, in conjunction with chronic social instability. Beginning 24 h after the first cat exposure, rats received daily injections of amitriptyline, clonidine, tianeptine or vehicle. Three weeks after the second cat exposure, all rats underwent a battery of behavioral and physiological tests. The vehicle-treated, psychosocially stressed rats demonstrated a robust fear memory for the two cat exposures, as well as increased anxiety expressed on the elevated plus maze, an exaggerated startle response, elevated heart rate and blood pressure, reduced growth rate and increased adrenal gland weight, relative to the vehicle-treated, non-stressed (control) rats. Neither amitriptyline nor clonidine was effective at blocking the entire cluster of stress-induced sequelae, and each agent produced adverse side effects in control subjects. Only the antidepressant tianeptine completely blocked the effects of psychosocial stress on all of the physiological and behavioral measures that were examined. These findings illustrate the differential effectiveness of these three treatments to block components of PTSD-like symptoms in rats, and in particular, reveal the profile of tianeptine as the most effective of all three agents.
Collapse
Affiliation(s)
- Phillip R Zoladz
- Department of Psychology, Sociology & Criminal Justice, Ohio Northern University, Ada, OH, USA
| | | | | |
Collapse
|
38
|
Perucca P, Mula M. Antiepileptic drug effects on mood and behavior: molecular targets. Epilepsy Behav 2013; 26:440-9. [PMID: 23092694 DOI: 10.1016/j.yebeh.2012.09.018] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Accepted: 09/06/2012] [Indexed: 01/09/2023]
Abstract
With almost 100 years of clinical experience, antiepileptic drugs (AEDs) remain the mainstay of epilepsy treatment. They suppress epileptic seizures by acting on a variety of mechanisms and molecular targets involved in the regulation of neuronal excitability. These include inhibitory-GABAergic and excitatory-glutamatergic neurotransmission, as well as ion (sodium and calcium) conductance through voltage-gated channels. On the other hand, accruing evidence indicates that these mechanisms and targets are also implicated in the regulation of mood and behavior, which may explain why each AED is associated with specific psychotropic effects. These effects, however, cannot be explained solely on the basis of the known mode of action of each AED, and other mechanisms or targets are likely to be implicated. In this article, we review positive and negative effects of AEDs on mood and behavior, discuss putative underlying mechanisms, and highlight knowledge gaps which should be addressed in future studies.
Collapse
Affiliation(s)
- Piero Perucca
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada.
| | | |
Collapse
|
39
|
Chandley M, Ordway G. Noradrenergic Dysfunction in Depression and Suicide. THE NEUROBIOLOGICAL BASIS OF SUICIDE 2012. [DOI: 10.1201/b12215-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
40
|
Levy BH, Tasker JG. Synaptic regulation of the hypothalamic-pituitary-adrenal axis and its modulation by glucocorticoids and stress. Front Cell Neurosci 2012; 6:24. [PMID: 22593735 PMCID: PMC3349941 DOI: 10.3389/fncel.2012.00024] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 04/26/2012] [Indexed: 12/12/2022] Open
Abstract
Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis has been implicated in a range of affective and stress-related disorders. The regulatory systems that control HPA activity are subject to modulation by environmental influences, and stressful life events or circumstances can promote subsequent HPA dysregulation. The brain is a major regulator of the HPA axis, and stress-induced plasticity of the neural circuitry involved in HPA regulation might constitute an etiological link between stress and the development of HPA dysregulation. This review focuses on the synaptic regulation of neuroendocrine corticotropin-releasing hormone (CRH) neurons of the hypothalamic paraventricular nucleus, which are the cells through which the brain predominantly exerts its influence on the HPA axis. CRH neuronal activity is largely orchestrated by three neurotransmitters: GABA, glutamate, and norepinephrine. We discuss our current understanding of the neural circuitry through which these neurotransmitters regulate CRH cell activity, as well as the plastic changes in this circuitry induced by acute and chronic stress and the resultant changes in HPA function.
Collapse
Affiliation(s)
| | - Jeffrey G. Tasker
- Neuroscience Program, Tulane University,New Orleans, LA, USA
- Department of Cell and Molecular Biology, Tulane University,New Orleans, LA, USA
| |
Collapse
|
41
|
Levy BH, Tasker JG. Synaptic regulation of the hypothalamic-pituitary-adrenal axis and its modulation by glucocorticoids and stress. Front Cell Neurosci 2012. [PMID: 22593735 DOI: 10.3389/fncel.2012.00024.ecollection] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis has been implicated in a range of affective and stress-related disorders. The regulatory systems that control HPA activity are subject to modulation by environmental influences, and stressful life events or circumstances can promote subsequent HPA dysregulation. The brain is a major regulator of the HPA axis, and stress-induced plasticity of the neural circuitry involved in HPA regulation might constitute an etiological link between stress and the development of HPA dysregulation. This review focuses on the synaptic regulation of neuroendocrine corticotropin-releasing hormone (CRH) neurons of the hypothalamic paraventricular nucleus, which are the cells through which the brain predominantly exerts its influence on the HPA axis. CRH neuronal activity is largely orchestrated by three neurotransmitters: GABA, glutamate, and norepinephrine. We discuss our current understanding of the neural circuitry through which these neurotransmitters regulate CRH cell activity, as well as the plastic changes in this circuitry induced by acute and chronic stress and the resultant changes in HPA function.
Collapse
Affiliation(s)
- Benjamin H Levy
- Neuroscience Program, Tulane University, New Orleans, LA, USA
| | | |
Collapse
|
42
|
Tse YC, Bagot RC, Hutter JA, Wong AS, Wong TP. Modulation of synaptic plasticity by stress hormone associates with plastic alteration of synaptic NMDA receptor in the adult hippocampus. PLoS One 2011; 6:e27215. [PMID: 22069501 PMCID: PMC3206081 DOI: 10.1371/journal.pone.0027215] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 10/12/2011] [Indexed: 01/22/2023] Open
Abstract
Stress exerts a profound impact on learning and memory, in part, through the actions of adrenal corticosterone (CORT) on synaptic plasticity, a cellular model of learning and memory. Increasing findings suggest that CORT exerts its impact on synaptic plasticity by altering the functional properties of glutamate receptors, which include changes in the motility and function of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid subtype of glutamate receptor (AMPAR) that are responsible for the expression of synaptic plasticity. Here we provide evidence that CORT could also regulate synaptic plasticity by modulating the function of synaptic N-methyl-D-aspartate receptors (NMDARs), which mediate the induction of synaptic plasticity. We found that stress level CORT applied to adult rat hippocampal slices potentiated evoked NMDAR-mediated synaptic responses within 30 min. Surprisingly, following this fast-onset change, we observed a slow-onset (>1 hour after termination of CORT exposure) increase in synaptic expression of GluN2A-containing NMDARs. To investigate the consequences of the distinct fast- and slow-onset modulation of NMDARs for synaptic plasticity, we examined the formation of long-term potentiation (LTP) and long-term depression (LTD) within relevant time windows. Paralleling the increased NMDAR function, both LTP and LTD were facilitated during CORT treatment. However, 1–2 hours after CORT treatment when synaptic expression of GluN2A-containing NMDARs is increased, bidirectional plasticity was no longer facilitated. Our findings reveal the remarkable plasticity of NMDARs in the adult hippocampus in response to CORT. CORT-mediated slow-onset increase in GluN2A in hippocampal synapses could be a homeostatic mechanism to normalize synaptic plasticity following fast-onset stress-induced facilitation.
Collapse
Affiliation(s)
- Yiu Chung Tse
- Neuroscience Division, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | - Rosemary C. Bagot
- Neuroscience Division, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | - Juliana A. Hutter
- Neuroscience Division, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | - Alice S. Wong
- Neuroscience Division, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | - Tak Pan Wong
- Neuroscience Division, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
43
|
Kennedy SH, Young AH, Blier P. Strategies to achieve clinical effectiveness: refining existing therapies and pursuing emerging targets. J Affect Disord 2011; 132 Suppl 1:S21-8. [PMID: 21571374 DOI: 10.1016/j.jad.2011.03.048] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 03/17/2011] [Indexed: 01/27/2023]
Abstract
BACKGROUND Clinical effectiveness reflects a balance between efficacy and tolerability as well as patient satisfaction and overall improvement in quality of life and function. This is of particular importance when considering the long term use of antidepressant therapies for relapse prevention. METHODS The purpose of this review is to explore methods to enhance the modest efficacy and effectiveness outcomes reported with current antidepressant strategies. Two strategies are addressed: a) Doing better with existing treatments and b) pursuing novel targets beyond the monoamine system for new antidepressant drug development. RESULTS In the first instance, it is important to consider the balance between antidepressant efficacy and tolerability for individual patients and also be aware of evidence supporting superiority of one agent over others. Both sequential and concurrent combination therapies with existing antidepressants are also reviewed. The second approach involves a review of emerging novel pharmacological treatments based on biomarker research. Unique targets where antidepressant treatments appear effective include the melatonergic, glutamatergic, neurotrophic, cytokine, and neuropeptide systems. CONCLUSIONS While agomelatine represents an example of a clinically available antidepressant that targets melatonin receptors, drugs that act on other candidate systems are still in the development phase.
Collapse
Affiliation(s)
- Sidney H Kennedy
- University Health Network, Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| | | | | |
Collapse
|
44
|
Beneficial Effects of Tianeptine on Hippocampus-Dependent Long-Term Memory and Stress-Induced Alterations of Brain Structure and Function. Pharmaceuticals (Basel) 2010. [PMCID: PMC4034085 DOI: 10.3390/ph3103143] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Tianeptine is a well-described antidepressant which has been shown to prevent stress from producing deleterious effects on brain structure and function. Preclinical studies have shown that tianeptine blocks stress-induced alterations of neuronal morphology and synaptic plasticity. Moreover, tianeptine prevents stress from impairing learning and memory, and, importantly, demonstrates memory-enhancing properties in the absence of stress. Recent research has indicated that tianeptine works by normalizing glutamatergic neurotransmission, a mechanism of action that may underlie its effectiveness as an antidepressant. These findings emphasize the value in focusing on the mechanisms of action of tianeptine, and specifically, the glutamatergic system, in the development of novel pharmacotherapeutic strategies in the treatment of depression.
Collapse
|
45
|
Stress-induced priming of glutamate synapses unmasks associative short-term plasticity. Nat Neurosci 2010; 13:1257-64. [PMID: 20818385 DOI: 10.1038/nn.2629] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 07/26/2010] [Indexed: 01/26/2023]
Abstract
Exposure to a stressor sensitizes or 'primes' the hypothalamic-pituitary-adrenal axis to a subsequent novel stressor. The synaptic mechanisms underlying this priming, however, are not known. We found that exposing a rat to a single stressor primed glutamate synapses in the paraventricular nucleus of the hypothalamus and allowed them to undergo a short-term potentiation (STP) following a burst of high-frequency afferent activity. This transient potentiation requires a corticotrophin-releasing hormone-dependent depression of postsynaptic NMDA receptors (NMDARs). The long-term depression of NMDAR function after stress prevented the vesicular release of an inhibitory retrograde messenger that, in control conditions, arrests STP. Following stress, STP manifested as an increase in the release probability of glutamate that was sufficient to induce multivesicular release. Our findings indicate that the priming of synapses to express STP is a synaptic correlate to stress-induced behavioral and neuroendocrine sensitization.
Collapse
|
46
|
Zoladz PR, Park CR, Muñoz C, Fleshner M, Diamond DM. Tianeptine: an antidepressant with memory-protective properties. Curr Neuropharmacol 2010; 6:311-21. [PMID: 19587852 PMCID: PMC2701287 DOI: 10.2174/157015908787386096] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 08/15/2008] [Accepted: 08/25/2008] [Indexed: 01/17/2023] Open
Abstract
The development of effective pharmacotherapy for major depression is important because it is such a widespread and debilitating mental disorder. Here, we have reviewed preclinical and clinical studies on tianeptine, an atypical antidepressant which ameliorates the adverse effects of stress on brain and memory. In animal studies, tianeptine has been shown to prevent stress-induced morphological sequelae in the hippocampus and amygdala, as well as to prevent stress from impairing synaptic plasticity in the prefrontal cortex and hippocampus. Tianeptine also has memory-protective characteristics, as it blocks the adverse effects of stress on hippocampus-dependent learning and memory. We have further extended the findings on stress, memory and tianeptine here with two novel observations: 1) stress impairs spatial memory in adrenalectomized (ADX), thereby corticosterone-depleted, rats; and 2) the stress-induced impairment of memory in ADX rats is blocked by tianeptine. These findings are consistent with previous research which indicates that tianeptine produces anti-stress and memory-protective properties without altering the response of the hypothalamic-pituitary-adrenal axis to stress. We conclude with a discussion of findings which indicate that tianeptine accomplishes its anti-stress effects by normalizing stress-induced increases in glutamate in the hippocampus and amygdala. This finding is potentially relevant to recent research which indicates that abnormalities in glutamatergic neurotransmission are involved in the pathogenesis of depression. Ultimately, tianeptine’s prevention of depression-induced sequelae in the brain is likely to be a primary factor in its effectiveness as a pharmacological treatment for depression.
Collapse
|
47
|
Effects of depressive-like behavior of rats on brain glutamate uptake. Neurochem Res 2010; 35:1164-71. [PMID: 20405205 DOI: 10.1007/s11064-010-0169-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2010] [Indexed: 12/30/2022]
Abstract
Learned helplessness paradigm is a widely accepted animal model of depressive-like behavior based on stress. Glutamatergic system is closely involved with the stress-neurotoxicity in the brain and recently it is pointed to have a relevant role in the pathophysiology of depression disorder. Glutamate uptake is the main mechanism to terminate the glutamatergic physiological activity and to neuroprotection against excitotoxicity. We investigated the profile of glutamate uptake in female rats submitted to the learned helplessness paradigm and to different classes of stress related to the paradigm, in slices of brain cortex, striatum and hippocampus. Glutamate uptake in slices of hippocampus differ between learned helplessness (LH) and non-learned helplessness (NLH) animals immediately persisting up to 21 days after the paradigm. In addition, there were a decrease of glutamate uptake in the three brain structures analyzed at 21 days after the paradigm for LH animals. These results may contribute to better understand the role of the glutamatergic system on the depressive-like behavior.
Collapse
|
48
|
|
49
|
Nakamichi N, Takarada T, Yoneda Y. Neurogenesis mediated by gamma-aminobutyric acid and glutamate signaling. J Pharmacol Sci 2009; 110:133-49. [PMID: 19483378 DOI: 10.1254/jphs.08r03cr] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
In this review, we will summarize our ongoing studies on the functionality of both gamma-aminobutyric acid (GABA) and glutamate receptors expressed by undifferentiated neural progenitor cells isolated from embryonic rodent brains. Cells were cultured with growth factors for the formation of round spheres by clustered cells under floating conditions, whereas a reverse transcription polymerase chain reaction analysis revealed expression of mRNA for particular subtypes of different ionotropic and metabotropic GABA and glutamate receptors in undifferentiated progenitors and neurospheres. Moreover, sustained exposure to either GABAergic or glutamatergic agonists not only modulated the size of neurospheres formed, but also affected spontaneous and induced differentiation of neural progenitor cells into particular progeny cell lineages such as neurons and astroglia. Both GABA and glutamate could play a pivotal role in the mechanisms underlying proliferation for self-replication along with the determination of subsequent differentiation fate toward particular progeny lineages through activation of their receptor subtypes functionally expressed by undifferentiated neural progenitor cells. Accordingly, neurogenesis seems to be also under control by GABAergic and glutamatergic signaling in developing brains as seen with neurotransmission in adult brains.
Collapse
Affiliation(s)
- Noritaka Nakamichi
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Natural Science and Technology, Ishikawa, Japan
| | | | | |
Collapse
|
50
|
Jalalvand E, Javan M, Haeri-Rohani A, Ahmadiani A. Stress- and non-stress-mediated mechanisms are involved in pain-induced apoptosis in hippocampus and dorsal lumbar spinal cord in rats. Neuroscience 2008; 157:446-52. [DOI: 10.1016/j.neuroscience.2008.08.052] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Revised: 08/23/2008] [Accepted: 08/26/2008] [Indexed: 12/12/2022]
|