1
|
Depoortère R, Papp M, Gruca P, Litwa E, Lason M, Biała D, Newman-Tancredi A. The antidepressant-like activity of ketamine in the rat chronic mild stress model requires activation of cortical 5-HT1A receptors. Behav Pharmacol 2024:00008877-990000000-00114. [PMID: 39718040 DOI: 10.1097/fbp.0000000000000809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
Ketamine displays efficacious rapid-acting antidepressant (RAAD) activity in the rat chronic mild stress (CMS) model. It rapidly reverses anhedonia (CMS-induced sucrose consumption deficit) and attenuates working memory deficit (novel object recognition: NOR) following both systemic (intraperitoneal, i.p.) administration or local administration in the prefrontal cortex (PFC). However, the receptor mechanisms underlying these effects remain to be clarified and may involve activation of serotonin 5-HT1A receptors, as previously found in experiments using the forced swim test. The present study explored the contribution of PFC 5-HT1A receptors in ketamine's RAAD activity in the CMS model. Ketamine (10 mg/kg i.p.) reversed CMS-induced sucrose consumption and working memory (NOR test) deficits. Notably, unilateral PFC microinjections of a 5-HT1A receptor antagonist, WAY-100635 (2 µg), prevented the antidepressant-like and pro-cognitive activity of systemic ketamine on sucrose consumption and working memory deficits. These data indicate that the RAAD activity of ketamine in the rat CMS model requires activation of PFC 5-HT1A receptors. They also reinforce the notion that drugs that directly activate PFC 5-HT1A receptors could constitute an alternative to ketamine as a promising strategy to achieve RAAD effects, with additional benefits against cognitive deficits in depressed patients, but without ketamine's troublesome side-effects and requirements for in-patient supervision.
Collapse
Affiliation(s)
| | - Mariusz Papp
- Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Piotr Gruca
- Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Ewa Litwa
- Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Magdalena Lason
- Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Dominika Biała
- Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | | |
Collapse
|
2
|
Wu A, Zhang J. Neuroinflammation, memory, and depression: new approaches to hippocampal neurogenesis. J Neuroinflammation 2023; 20:283. [PMID: 38012702 PMCID: PMC10683283 DOI: 10.1186/s12974-023-02964-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023] Open
Abstract
As one of most common and severe mental disorders, major depressive disorder (MDD) significantly increases the risks of premature death and other medical conditions for patients. Neuroinflammation is the abnormal immune response in the brain, and its correlation with MDD is receiving increasing attention. Neuroinflammation has been reported to be involved in MDD through distinct neurobiological mechanisms, among which the dysregulation of neurogenesis in the dentate gyrus (DG) of the hippocampus (HPC) is receiving increasing attention. The DG of the hippocampus is one of two niches for neurogenesis in the adult mammalian brain, and neurotrophic factors are fundamental regulators of this neurogenesis process. The reported cell types involved in mediating neuroinflammation include microglia, astrocytes, oligodendrocytes, meningeal leukocytes, and peripheral immune cells which selectively penetrate the blood-brain barrier and infiltrate into inflammatory regions. This review summarizes the functions of the hippocampus affected by neuroinflammation during MDD progression and the corresponding influences on the memory of MDD patients and model animals.
Collapse
Affiliation(s)
- Anbiao Wu
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Jiyan Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
| |
Collapse
|
3
|
van Hagen BTJ, van Goethem NP, Nelissen E, Paes D, Koymans K, van Hoof S, Schreiber R, Varney M, Newman-Tancredi A, Prickaerts J. Biased 5-HT 1A receptor agonists F13714 and NLX-101 differentially affect pattern separation and neuronal plasticity in rats after acute and chronic treatment. Mol Cell Neurosci 2022; 120:103719. [PMID: 35283305 DOI: 10.1016/j.mcn.2022.103719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/16/2022] [Accepted: 03/07/2022] [Indexed: 11/28/2022] Open
Abstract
Pattern separation is a hippocampal process in which highly similar stimuli are recognized as separate representations, and deficits could lead to memory impairments in neuropsychiatric disorders such as schizophrenia. The 5-HT1A receptor (5-HT1AR) is believed to be involved in these hippocampal pattern separation processes. However, in the dorsal raphe nucleus (DRN), the 5-HT1AR is expressed as a somatodendritic autoreceptor, negatively regulates serotonergic signaling, and could thereby counteract the effects of hippocampal postsynaptic 5-HT1A receptors. Therefore, this study aims to identify how pre- and post-synaptic 5-HT1AR activity affects pattern separation. Object pattern separation (OPS) performance was measured in male Wistar rats after both acute and chronic treatment (i.p.) with 5-HT1AR biased agonists F13714 (0.0025 mg/kg acutely, 0.02 mg/kg/day chronically) or NLX-101 (0.08 mg/kg acutely, 0.32 mg/kg/day chronically), which preferentially activate autoreceptors or postsynaptic receptors respectively, for 14 days. Body temperature - a functional correlate of hypothalamic 5-HT1AR stimulation - was measured daily. Additionally, 5-HT1AR density (DRN) and plasticity markers (hippocampus) were assessed. Acute treatment with F13714 impaired OPS performance, whereas chronic treatment normalized this, and a drop in body temperature was found from day 4 onwards. NLX-101 enhanced OPS performance acutely and chronically, and caused an acute drop in body temperature. Chronic NLX-101 treatment increased doublecortin positive neurons in the dorsal hippocampus, while chronic treatment with F13714 resulted in a downregulation of 5-HT1A autoreceptors, which likely reversed the acute impairment in OPS performance. Chronic treatment with NLX-101 appears to have therapeutic potential to improve brain plasticity and OPS performance.
Collapse
Affiliation(s)
- Britt T J van Hagen
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Nick P van Goethem
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Ellis Nelissen
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Dean Paes
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Karin Koymans
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands; Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, the Netherlands
| | - Scott van Hoof
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands; German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany
| | - Rudy Schreiber
- Faculty of Psychology and Neuroscience, Section Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, the Netherlands
| | - Mark Varney
- Neurolixis Inc., Dana Point, California, USA
| | | | - Jos Prickaerts
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
4
|
Leschik J, Lutz B, Gentile A. Stress-Related Dysfunction of Adult Hippocampal Neurogenesis-An Attempt for Understanding Resilience? Int J Mol Sci 2021; 22:7339. [PMID: 34298958 PMCID: PMC8305135 DOI: 10.3390/ijms22147339] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/16/2022] Open
Abstract
Newborn neurons in the adult hippocampus are regulated by many intrinsic and extrinsic cues. It is well accepted that elevated glucocorticoid levels lead to downregulation of adult neurogenesis, which this review discusses as one reason why psychiatric diseases, such as major depression, develop after long-term stress exposure. In reverse, adult neurogenesis has been suggested to protect against stress-induced major depression, and hence, could serve as a resilience mechanism. In this review, we will summarize current knowledge about the functional relation of adult neurogenesis and stress in health and disease. A special focus will lie on the mechanisms underlying the cascades of events from prolonged high glucocorticoid concentrations to reduced numbers of newborn neurons. In addition to neurotransmitter and neurotrophic factor dysregulation, these mechanisms include immunomodulatory pathways, as well as microbiota changes influencing the gut-brain axis. Finally, we discuss recent findings delineating the role of adult neurogenesis in stress resilience.
Collapse
Affiliation(s)
- Julia Leschik
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany;
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany;
- Leibniz Institute for Resilience Research (LIR), 55122 Mainz, Germany
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy;
| |
Collapse
|
5
|
Jorgensen C, Wang Z. Hormonal Regulation of Mammalian Adult Neurogenesis: A Multifaceted Mechanism. Biomolecules 2020; 10:biom10081151. [PMID: 32781670 PMCID: PMC7465680 DOI: 10.3390/biom10081151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/27/2020] [Accepted: 08/01/2020] [Indexed: 02/07/2023] Open
Abstract
Adult neurogenesis—resulting in adult-generated functioning, integrated neurons—is still one of the most captivating research areas of neuroplasticity. The addition of new neurons in adulthood follows a seemingly consistent multi-step process. These neurogenic stages include proliferation, differentiation, migration, maturation/survival, and integration of new neurons into the existing neuronal network. Most studies assessing the impact of exogenous (e.g., restraint stress) or endogenous (e.g., neurotrophins) factors on adult neurogenesis have focused on proliferation, survival, and neuronal differentiation. This review will discuss the multifaceted impact of hormones on these various stages of adult neurogenesis. Specifically, we will review the evidence for hormonal facilitation (via gonadal hormones), inhibition (via glucocorticoids), and neuroprotection (via recruitment of other neurochemicals such as neurotrophin and neuromodulators) on newly adult-generated neurons in the mammalian brain.
Collapse
Affiliation(s)
- Claudia Jorgensen
- Behavioral Science Department, Utah Valley University, Orem, UT 84058, USA
- Correspondence:
| | - Zuoxin Wang
- Psychology Department and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA;
| |
Collapse
|
6
|
Foltran RB, Stefani KM, Bonafina A, Resasco A, Diaz SL. Differential Hippocampal Expression of BDNF Isoforms and Their Receptors Under Diverse Configurations of the Serotonergic System in a Mice Model of Increased Neuronal Survival. Front Cell Neurosci 2019; 13:384. [PMID: 31555094 PMCID: PMC6712164 DOI: 10.3389/fncel.2019.00384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 08/06/2019] [Indexed: 12/14/2022] Open
Abstract
Neurotrophic factors are relevant regulators of the neurogenic process at different levels. In particular, the brain-derived neurotrophic factor, BDNF, is highly expressed in the hippocampus (HC) of rodents and participates in the control of neuronal proliferation, and survival in the dentate gyrus (DG). Likewise, serotonin is also involved in the regulation of neurogenesis, though its role is apparently more complex. Indeed, both enhancement of serotonin neurotransmission as well as serotonin depletion, paradoxically increase neuronal survival in the HC of mice. In this study, we analyzed the protein expression of the BDNF isoforms, i.e., pro- and mature-BDNF, and their respective receptors p75 and TrkB, in the HC of mice chronically treated with para-chloro-phenyl-alanine (PCPA), an inhibitor of serotonin synthesis. The same analysis was conducted in hyposerotonergic mice with concomitant administration of the 5-HT1A receptor agonist, 8-Hydroxy-2-(di-n- propylamino) tetralin (8-OH-DPAT). Increased expression of p75 receptor with decreased expression of pro-BDNF was observed after chronic PCPA. Seven-day treatment with 8-OH-DPAT reestablished the expression of pro-BDNF modified by PCPA, and induced an increase in the expression of p75 receptor. It has been demonstrated that PCPA-treated mice have higher number of immature neurons in the HC. Given that immature neurons participate in the pattern separation process, the object pattern separation test was conducted. A better performance of hyposerotonergic mice was not confirmed in this assay. Altogether, our results show that molecules in the BDNF signaling pathway are differentially expressed under diverse configurations of the serotonergic system, allowing for fine-tuning of the neurogenic process.
Collapse
Affiliation(s)
- Rocío Beatriz Foltran
- Instituto de Biología Celular y Neurociencia Prof. E. De Robertis, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Karen Melany Stefani
- Instituto de Biología Celular y Neurociencia Prof. E. De Robertis, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Antonela Bonafina
- Instituto de Biología Celular y Neurociencia Prof. E. De Robertis, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Agustina Resasco
- Instituto de Biología Celular y Neurociencia Prof. E. De Robertis, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Silvina Laura Diaz
- Instituto de Biología Celular y Neurociencia Prof. E. De Robertis, CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
7
|
Tert-butyl 4-((1-phenyl-1H-pyrazol-4-yl) methyl) piperazine-1-carboxylate (LQFM104)– New piperazine derivative with antianxiety and antidepressant-like effects: Putative role of serotonergic system. Biomed Pharmacother 2018; 103:546-552. [DOI: 10.1016/j.biopha.2018.04.077] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 01/22/2023] Open
|
8
|
Godoy LD, Umeoka EHL, Ribeiro DE, Santos VR, Antunes-Rodrigues J, Joca SRL, Garcia-Cairasco N. Multimodal early-life stress induces biological changes associated to psychopathologies. Horm Behav 2018; 100:69-80. [PMID: 29548783 DOI: 10.1016/j.yhbeh.2018.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 03/07/2018] [Accepted: 03/11/2018] [Indexed: 01/21/2023]
Abstract
Evidences suggest the contributive role of early-life stress (ELS) to affective and anxiety disorders. Chronic exposure to the same stressor may generate habituation, while the exposure to different and repeated stressors gradually promotes maladaptive plasticity. Therefore, to further understand the effects of heterotypic stressors during early life period, male Wistar rat pups (P1-P21) were exposed to Multimodal ELS paradigm. Results indicate pups did not habituate to multimodal ELS and neonates respond to both physical and psychogenic stressors. Adult rats that underwent ELS protocol showed significant lower sucrose intake, decreased latency to immobility in the forced swim test and increased latency to light compartment in the light-dark test when compared to control group. Although it has been shown that ELS-induced changes in hippocampus can be used as biomarkers, multimodal ELS did not significantly alter BDNF, Tyrosine Kinase B (TrkB) receptor expression or neurogenesis in the hippocampus. Taken together, these findings indicate that multimodal ELS protocol can be an interesting experimental model for understanding long-term psychiatric disorders associated with stress. Indeed, our data with neurogenesis, BDNF and TrkB, and conflicting data from the literature, suggest that additional studies on synaptic plasticity/intracellular cascades would help to detect the underlying mechanisms.
Collapse
Affiliation(s)
- Lívea Dornela Godoy
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | - Eduardo H L Umeoka
- Neurosciences and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | - Deidiane Elisa Ribeiro
- Pharmacology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | | | - José Antunes-Rodrigues
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | - Samia Regiane Lourenço Joca
- Physics and Chemistry Department, Ribeirão Preto School of Pharmacy, University of São Paulo, Brazil; Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
| | - Norberto Garcia-Cairasco
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil; Neurosciences and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil.
| |
Collapse
|
9
|
Abstract
Previous attempts to identify a unified theory of brain serotonin function have largely failed to achieve consensus. In this present synthesis, we integrate previous perspectives with new and older data to create a novel bipartite model centred on the view that serotonin neurotransmission enhances two distinct adaptive responses to adversity, mediated in large part by its two most prevalent and researched brain receptors: the 5-HT1A and 5-HT2A receptors. We propose that passive coping (i.e. tolerating a source of stress) is mediated by postsynaptic 5-HT1AR signalling and characterised by stress moderation. Conversely, we argue that active coping (i.e. actively addressing a source of stress) is mediated by 5-HT2AR signalling and characterised by enhanced plasticity (defined as capacity for change). We propose that 5-HT1AR-mediated stress moderation may be the brain's default response to adversity but that an improved ability to change one's situation and/or relationship to it via 5-HT2AR-mediated plasticity may also be important - and increasingly so as the level of adversity reaches a critical point. We propose that the 5-HT1AR pathway is enhanced by conventional 5-HT reuptake blocking antidepressants such as the selective serotonin reuptake inhibitors (SSRIs), whereas the 5-HT2AR pathway is enhanced by 5-HT2AR-agonist psychedelics. This bipartite model purports to explain how different drugs (SSRIs and psychedelics) that modulate the serotonergic system in different ways, can achieve complementary adaptive and potentially therapeutic outcomes.
Collapse
Affiliation(s)
- RL Carhart-Harris
- Psychedelic Research Group, Neuropsychopharmacology Unit, Centre for Psychiatry, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - DJ Nutt
- Psychedelic Research Group, Neuropsychopharmacology Unit, Centre for Psychiatry, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
10
|
Fitzsimons CP, Herbert J, Schouten M, Meijer OC, Lucassen PJ, Lightman S. Circadian and ultradian glucocorticoid rhythmicity: Implications for the effects of glucocorticoids on neural stem cells and adult hippocampal neurogenesis. Front Neuroendocrinol 2016; 41:44-58. [PMID: 27234350 DOI: 10.1016/j.yfrne.2016.05.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 05/01/2016] [Accepted: 05/03/2016] [Indexed: 01/01/2023]
Abstract
Psychosocial stress, and within the neuroendocrine reaction to stress specifically the glucocorticoid hormones, are well-characterized inhibitors of neural stem/progenitor cell proliferation in the adult hippocampus, resulting in a marked reduction in the production of new neurons in this brain area relevant for learning and memory. However, the mechanisms by which stress, and particularly glucocorticoids, inhibit neural stem/progenitor cell proliferation remain unclear and under debate. Here we review the literature on the topic and discuss the evidence for direct and indirect effects of glucocorticoids on neural stem/progenitor cell proliferation and adult neurogenesis. Further, we discuss the hypothesis that glucocorticoid rhythmicity and oscillations originating from the activity of the hypothalamus-pituitary-adrenal axis, may be crucial for the regulation of neural stem/progenitor cells in the hippocampus, as well as the implications of this hypothesis for pathophysiological conditions in which glucocorticoid oscillations are affected.
Collapse
Affiliation(s)
- Carlos P Fitzsimons
- Neuroscience Program, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098XH Amsterdam, The Netherlands.
| | - Joe Herbert
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Marijn Schouten
- Neuroscience Program, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098XH Amsterdam, The Netherlands
| | - Onno C Meijer
- Leiden University Medical Centre, Department of Endocrinology, Leiden, The Netherlands
| | - Paul J Lucassen
- Neuroscience Program, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098XH Amsterdam, The Netherlands.
| | - Stafford Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, United Kingdom
| |
Collapse
|
11
|
Dogra S, Sona C, Kumar A, Yadav PN. Epigenetic regulation of G protein coupled receptor signaling and its implications in psychiatric disorders. Int J Biochem Cell Biol 2016; 77:226-39. [PMID: 27046448 DOI: 10.1016/j.biocel.2016.03.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 12/21/2022]
Abstract
G protein-coupled receptors (GPCRs) act as a relay center through which extracellular signals, in the form of neurotransmitters or therapeutics, are converted into an intracellular response, which ultimately shapes the overall response at the tissue and behavioral level. Remarkably in similar ways, epigenetic mechanisms also modulate the expression pattern of a large number of genes in response to the dynamic environment inside and outside of the body, and consequently overall response. Emerging evidences from the pharmacogenomics and preclinical studies clearly suggest that these two distinct mechanisms criss-cross each other in several neurological disorders. At one hand such cross-talks between two distinct mechanisms make disease etiology more challenging to understand, while on the other hand if dealt appropriately, such situations might provide an opportunity to find novel druggable target and strategy for the treatment of complex diseases. In this review article, we have summarized and highlighted the main findings that tie epigenetic mechanisms to GPCR mediated signaling in the pathophysiology of central nervous system (CNS) disorders, including depression, addiction and pain.
Collapse
Affiliation(s)
- Shalini Dogra
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Chandan Sona
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Ajeet Kumar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Prem N Yadav
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India.
| |
Collapse
|
12
|
Qesseveur G, Petit AC, Nguyen HT, Dahan L, Colle R, Rotenberg S, Seif I, Robert P, David D, Guilloux JP, Gardier AM, Verstuyft C, Becquemont L, Corruble E, Guiard BP. Genetic dysfunction of serotonin 2A receptor hampers response to antidepressant drugs: A translational approach. Neuropharmacology 2016; 105:142-153. [PMID: 26764241 DOI: 10.1016/j.neuropharm.2015.12.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/23/2015] [Accepted: 12/24/2015] [Indexed: 01/31/2023]
Abstract
Pharmacological studies have yielded valuable insights into the role of the serotonin 2A (5-HT2A) receptor in major depressive disorder (MDD) and antidepressant drugs (ADs) response. However, it is still unknown whether genetic variants in the HTR2A gene affect the therapeutic outcome of ADs and the mechanism underlying the regulation of such response remains poorly described. In this context, a translational human-mouse study offers a unique opportunity to address the possibility that variations in the HTR2A gene may represent a relevant marker to predict the efficacy of ADs. In a first part of this study, we investigated in depressed patients the effect of three HTR2A single nucleotide polymorphisms (SNPs), selected for their potential functional consequences on 5-HT2A receptor (rs6313, rs6314 and rs7333412), on response and remission rates after 3 months of antidepressant treatments. We also explored the consequences of the constitutive genetic inactivation of the 5-HT2A receptor (i.e. in 5-HT2A(-/-) mice) on the activity of acute and prolonged administration of SSRIs. Our clinical data indicate that GG patients for the rs7333412 SNP were less prone to respond to ADs than AA/AG patients. In the preclinical study, we demonstrated that the 5-HT2A receptor exerts an inhibitory influence on the neuronal activity of the serotonergic system after acute administration of SSRIs. However, while the chronic administration of the SSRIs escitalopram or fluoxetine elicited a progressive increased in the firing rate of 5-HT neurons in 5-HT2A(+/+) mice, it failed to do so in 5-HT2A(-/-) mutants. These electrophysiological impairments were associated with a decreased ability of the chronic administration of fluoxetine to stimulate hippocampal plasticity and to produce antidepressant-like activities. Genetic loss of the 5-HT2A receptor compromised the activity of chronic treatment with SSRIs, making this receptor a putative marker to predict ADs response.
Collapse
Affiliation(s)
- Gaël Qesseveur
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, Fac Pharmacie, Châtenay Malabry, 92290, France
| | - Anne Cécile Petit
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, CESP, Fac Médecine Paris Sud, 94275, Le Kremlin Bicêtre, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital de Bicêtre, Service de Psychiatrie, Le Kremlin Bicêtre, F-94275, France
| | - Hai Thanh Nguyen
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, Fac Pharmacie, Châtenay Malabry, 92290, France
| | - Lionel Dahan
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| | - Romain Colle
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, CESP, Fac Médecine Paris Sud, 94275, Le Kremlin Bicêtre, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital de Bicêtre, Service de Psychiatrie, Le Kremlin Bicêtre, F-94275, France
| | - Samuel Rotenberg
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, CESP, Fac Médecine Paris Sud, 94275, Le Kremlin Bicêtre, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital de Bicêtre, Service de Psychiatrie, Le Kremlin Bicêtre, F-94275, France
| | - Isabelle Seif
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, Fac Pharmacie, Châtenay Malabry, 92290, France
| | - Pauline Robert
- UMS IPSIT (INST. Paris-Saclay d'innovation Thérapeutique), Paris Sud, France
| | - Denis David
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, Fac Pharmacie, Châtenay Malabry, 92290, France
| | - Jean-Philippe Guilloux
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, Fac Pharmacie, Châtenay Malabry, 92290, France
| | - Alain M Gardier
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, Fac Pharmacie, Châtenay Malabry, 92290, France
| | - Céline Verstuyft
- INSERM U1184, Le Kremlin Bicêtre, F-94276, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital de Bicêtre, Service de Génétique moléculaire, Pharmacogénétique et Hormonologie, Le Kremlin Bicêtre, F-94275, France
| | - Laurent Becquemont
- INSERM U1184, Le Kremlin Bicêtre, F-94276, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital de Bicêtre, Service de Génétique moléculaire, Pharmacogénétique et Hormonologie, Le Kremlin Bicêtre, F-94275, France
| | - Emmanuelle Corruble
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, CESP, Fac Médecine Paris Sud, 94275, Le Kremlin Bicêtre, France; Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital de Bicêtre, Service de Psychiatrie, Le Kremlin Bicêtre, F-94275, France.
| | - Bruno P Guiard
- Université Paris-Saclay, Univ. Paris-Sud, INSERM UMR-S 1178, Fac Pharmacie, Châtenay Malabry, 92290, France; Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| |
Collapse
|
13
|
Lu KT, Huang TC, Wang JY, You YS, Chou JL, Chan MWY, Wo PYY, Amstislavskaya TG, Tikhonova MA, Yang YL. NKCC1 mediates traumatic brain injury-induced hippocampal neurogenesis through CREB phosphorylation and HIF-1α expression. Pflugers Arch 2015; 467:1651-61. [PMID: 25201604 DOI: 10.1007/s00424-014-1588-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 07/08/2014] [Accepted: 07/28/2014] [Indexed: 01/03/2023]
Abstract
Traumatic brain injury (TBI) is one of the most prevalent causes of worldwide mortality and morbidity. We previously had evidenced that TBI induced Na-K-2Cl co-transporter (NKCC1) upregulation in hippocampus. Here, we aim to investigate the role of NKCC1 in TBI-induced neurogenesis and the detailed mechanisms. The TBI-associated alternations in the expression of NKCC1, HIF-1α, VEGF, MAPK cascade, and CREB phosphorylation were analyzed by Western blot. TBI-induced neurogenesis was determined by immuno-fluorescence labeling. Chromatin immunoprecipitation was used to elucidate whether HIF-1α would activate VEGF gene after TBI. We found that the level of hippocampal NKCC1 and VEGF began to rise 8 h after TBI, and both of them reached maxima at day 7. Along with the upregulation of NKCC1 and VEGF, MAPK cascade was activated and hippocampal neurogenesis was promoted. Administration of CREB antisense oligonucleotide significantly attenuated the expression of HIF-1α, while HIF-1α antisense oligonucleotide exhibited little effect on the expression of CREB. However, HIF-1α antisense oligonucleotide administration did effectively suppress the expression of VEGF. Our results of the chromosome immunoprecipitation also indicated that HIF-1α could directly act on the VEGF promoter and presumably would elevate the VEGF expression after TBI. All these results have illustrated the correlation between NKCC1 upregulation and TBI-associated neurogenesis. The pathway involves the activation of Raf/MEK/ERK cascade, CREB phosphorylation, and HIF-1α upregulation, and finally leads to the stimulation of VEGF expression and the induction of neurogenesis.
Collapse
Affiliation(s)
- Kwok-Tung Lu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Murata Y, Yanagihara Y, Mori M, Mine K, Enjoji M. Chronic treatment with tandospirone, a serotonin 1A receptor partial agonist, inhibits psychosocial stress-induced changes in hippocampal neurogenesis and behavior. J Affect Disord 2015; 180:1-9. [PMID: 25879718 DOI: 10.1016/j.jad.2015.03.054] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 03/26/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND The 5-hydroxytryptamine (5-HT) 1A receptors are considered a potential target for the treatment of mental and neuropsychiatric disorders. Several studies have indicated that 5-HT1A receptor agonists increase hippocampal neurogenesis, which is implicated in the action mechanism of antidepressants. However, these agents have not been applied to humans due to intolerable side effects. We recently showed that chronic administration of tandospirone, a clinically available 5-HT1A receptor partial agonist, increased hippocampal neurogenesis dose-dependently. The present study was done to determine if chronic tandospirone treatment has antidepressant potential from the standpoint of hippocampal neurogenesis and behavior. METHODS Male Sprague-Dawley rats were intraperitoneally administered a vehicle or tandospirone (10mg/kg) once daily for 28 days. Two weeks after starting the injections, animals were exposed to intermittent social defeat (four times over two weeks). The effects of stress and tandospirone on the rodents׳ behavior were evaluated by the Novelty-Suppressed Feeding (NSF) test. The quantification of hippocampal neurogenesis was estimated using immunostaining with Ki-67 and doublecortin (DCX). RESULTS Chronic tandospirone treatment reversed the psychosocial stress-induced increase in the latency in the NSF test and decrease in the density of DCX-positive cells in the dentate gyrus of the dorsal and ventral hippocampus. However, no difference in the density of Ki-67-positive cells was observed between the vehicle- and tandospirone-administered groups. LIMITATIONS To clarify the antidepressant potential of TDS, the other behavioral tests for depression will be required. CONCLUSIONS Our findings suggest that tandospirone has antidepressant potential through an inhibiting effect on stress-induced changes in hippocampal neurogenesis.
Collapse
Affiliation(s)
- Yusuke Murata
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Yuki Yanagihara
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Masayoshi Mori
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Kazunori Mine
- Faculty of Neurology and Psychiatry, Mito Hospital, 4-1-1, Shime-Higashi, Shime-Machi, Kasuya-Gun, Fukuoka 811-2243, Japan
| | - Munechika Enjoji
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| |
Collapse
|
15
|
Imoto Y, Kira T, Sukeno M, Nishitani N, Nagayasu K, Nakagawa T, Kaneko S, Kobayashi K, Segi-Nishida E. Role of the 5-HT4 receptor in chronic fluoxetine treatment-induced neurogenic activity and granule cell dematuration in the dentate gyrus. Mol Brain 2015; 8:29. [PMID: 25976618 PMCID: PMC4430984 DOI: 10.1186/s13041-015-0120-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/24/2015] [Indexed: 11/30/2022] Open
Abstract
Background Chronic treatment with selective serotonin (5-HT) reuptake inhibitors (SSRIs) facilitates adult neurogenesis and reverses the state of maturation in mature granule cells (GCs) in the dentate gyrus (DG) of the hippocampus. Recent studies have suggested that the 5-HT4 receptor is involved in both effects. However, it is largely unknown how the 5-HT4 receptor mediates neurogenic effects in the DG and, how the neurogenic and dematuration effects of SSRIs interact with each other. Results We addressed these issues using 5-HT4 receptor knockout (5-HT4R KO) mice. Expression of the 5-HT4 receptor was detected in mature GCs but not in neuronal progenitors of the DG. We found that chronic treatment with the SSRI fluoxetine significantly increased cell proliferation and the number of doublecortin-positive cells in the DG of wild-type mice, but not in 5-HT4R KO mice. We then examined the correlation between the increased neurogenesis and the dematuration of GCs. As reported previously, reduced expression of calbindin in the DG, as an index of dematuration, by chronic fluoxetine treatment was observed in wild-type mice but not in 5-HT4R KO mice. The proliferative effect of fluoxetine was inversely correlated with the expression level of calbindin in the DG. The expression of neurogenic factors in the DG, such as brain derived neurotrophic factor (Bdnf), was also associated with the progression of dematuration. These results indicate that the neurogenic effects of fluoxetine in the DG are closely associated with the progression of dematuration of GCs. In contrast, the DG in which neurogenesis was impaired by irradiation still showed significant reduction of calbindin expression by chronic fluoxetine treatment, suggesting that dematuration of GCs by fluoxetine does not require adult neurogenesis in the DG. Conclusions We demonstrated that the 5-HT4 receptor plays an important role in fluoxetine-induced adult neurogenesis in the DG in addition to GC dematuration, and that these phenomena are closely associated. Our results suggest that 5-HT4 receptor-mediated phenotypic changes, including dematuration in mature GCs, underlie the neurogenic effect of SSRIs in the DG, providing new insight into the cellular mechanisms of the neurogenic actions of SSRIs in the hippocampus.
Collapse
Affiliation(s)
- Yuki Imoto
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences Kyoto University, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Toshihiko Kira
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences Kyoto University, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Mamiko Sukeno
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences Kyoto University, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Naoya Nishitani
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences Kyoto University, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences Kyoto University, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Takayuki Nakagawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences Kyoto University, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences Kyoto University, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Katsunori Kobayashi
- Department of Pharmacology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan. .,Japan Science and Technology Agency, Core Research for Evolutional Science and Technology, Saitama, 332-0012, Japan.
| | - Eri Segi-Nishida
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan.
| |
Collapse
|
16
|
Hui J, Zhang J, Kim H, Tong C, Ying Q, Li Z, Mao X, Shi G, Yan J, Zhang Z, Xi G. Fluoxetine regulates neurogenesis in vitro through modulation of GSK-3β/β-catenin signaling. Int J Neuropsychopharmacol 2015; 18:pyu099. [PMID: 25522429 PMCID: PMC4376550 DOI: 10.1093/ijnp/pyu099] [Citation(s) in RCA: 47] [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] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND It is generally accepted that chronic treatment with antidepressants increases hippocampal neurogenesis, but the molecular mechanisms underlying their effects are unknown. Recently, glycogen synthase kinase-3 beta (GSK-3β)/β-catenin signaling was shown to be involved in the mechanism of how antidepressants might influence hippocampal neurogenesis. METHODS The aim of this study was to determine whether GSK-3β/β-catenin signaling is involved in the alteration of neurogenesis as a result of treatment with fluoxetine, a selective serotonin reuptake inhibitor. The mechanisms involved in fluoxetine's regulation of GSK-3β/β-catenin signaling pathway were also examined. RESULTS Our results demonstrated that fluoxetine increased the proliferation of embryonic neural precursor cells (NPCs) by up-regulating the phosphorylation of Ser9 on GSK-3β and increasing the level of nuclear β-catenin. The overexpression of a stabilized β-catenin protein (ΔN89 β-catenin) significantly increased NPC proliferation, while inhibition of β-catenin expression in NPCs led to a significant decrease in the proliferation and reduced the proliferative effects induced by fluoxetine. The effects of fluoxetine-induced up-regulation of both phosphorylation of Ser9 on GSK-3β and nuclear β-catenin were significantly prevented by the 5-hydroxytryptamine-1A (5-HT1A) receptor antagonist WAY-100635. CONCLUSIONS The results demonstrate that fluoxetine may increase neurogenesis via the GSK-3β/β-catenin signaling pathway that links postsynaptic 5-HT1A receptor activation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Guangjun Xi
- Department of Critical Care Medicine, Wuxi People's Hospital of Nanjing Medical University, Wuxi, China (Drs Hui and Yan); Department of Neurology, Wuxi People's Hospital of Nanjing Medical University, Wuxi, China (Drs J Zhang, Li, Mao, Shi, and Xi); Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Cell and Neurobiology, University of Southern California, Los Angeles, CA (Drs Kim, Tong, and Ying); Department of Neurology, Affiliated ZhongDa Hospital of Southeast University, Nanjing, China (Dr Z Zhang).
| |
Collapse
|
17
|
The role of serotonin in adult hippocampal neurogenesis. Behav Brain Res 2014; 277:49-57. [PMID: 25125239 DOI: 10.1016/j.bbr.2014.07.038] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/22/2014] [Accepted: 07/23/2014] [Indexed: 12/17/2022]
Abstract
Serotonin is probably best known for its role in conveying a sense of contentedness and happiness. It is one of the most unique and pharmacologically complex monoamines in both the peripheral and central nervous system (CNS). Serotonin has become in focus of interest for the treatment of depression with multiple serotonin-mimetic and modulators of adult neurogenesis used clinically. Here we will take a broad view of serotonin from development to its physiological role as a neurotransmitter and its contribution to homeostasis of the adult rodent hippocampus. This chapter reflects the most significant findings on cellular and molecular mechanisms from neuroscientists in the field over the last two decades. We illustrate the action of serotonin by highlighting basic receptor targeting studies, and how receptors impact brain function. We give an overview of recent genetically modified mouse models that differ in serotonin availability and focus on the role of the monoamine in antidepressant response. We conclude with a synthesis of the most recent data surrounding the role of serotonin in activity and hippocampal neurogenesis. This synopsis sheds light on the mechanisms and potential therapeutic model by which serotonin plays a critical role in the maintenance of mood.
Collapse
|
18
|
Islam M, Moriguchi S, Tagashira H, Fukunaga K. Rivastigmine improves hippocampal neurogenesis and depression-like behaviors via 5-HT1A receptor stimulation in olfactory bulbectomized mice. Neuroscience 2014; 272:116-30. [DOI: 10.1016/j.neuroscience.2014.04.046] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 04/23/2014] [Accepted: 04/23/2014] [Indexed: 11/29/2022]
|
19
|
Coplan JD, Gopinath S, Abdallah CG, Berry BR. A neurobiological hypothesis of treatment-resistant depression - mechanisms for selective serotonin reuptake inhibitor non-efficacy. Front Behav Neurosci 2014; 8:189. [PMID: 24904340 PMCID: PMC4033019 DOI: 10.3389/fnbeh.2014.00189] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/07/2014] [Indexed: 12/20/2022] Open
Abstract
First-line treatment of major depression includes administration of a selective serotonin reuptake inhibitor (SSRI), yet studies suggest that remission rates following two trials of an SSRI are <50%. The authors examine the putative biological substrates underlying "treatment resistant depression (TRD)" with the goal of elucidating novel rationales to treat TRD. We look at relevant articles from the preclinical and clinical literature combined with clinical exposure to TRD patients. A major focus was to outline pathophysiological mechanisms whereby the serotonin system becomes impervious to the desired enhancement of serotonin neurotransmission by SSRIs. A complementary focus was to dissect neurotransmitter systems, which serve to inhibit the dorsal raphe. We propose, based on a body of translational studies, TRD may not represent a simple serotonin deficit state but rather an excess of midbrain peri-raphe serotonin and subsequent deficit at key fronto-limbic projection sites, with ultimate compromise in serotonin-mediated neuroplasticity. Glutamate, serotonin, noradrenaline, and histamine are activated by stress and exert an inhibitory effect on serotonin outflow, in part by "flooding" 5-HT1A autoreceptors by serotonin itself. Certain factors putatively exacerbate this scenario - presence of the short arm of the serotonin transporter gene, early-life adversity and comorbid bipolar disorder - each of which has been associated with SSRI-treatment resistance. By utilizing an incremental approach, we provide a system for treating the TRD patient based on a strategy of rescuing serotonin neurotransmission from a state of SSRI-induced dorsal raphe stasis. This calls for "stacked" interventions, with an SSRI base, targeting, if necessary, the glutamatergic, serotonergic, noradrenergic, and histaminergic systems, thereby successively eliminating the inhibitory effects each are capable of exerting on serotonin neurons. Future studies are recommended to test this biologically based approach for treatment of TRD.
Collapse
Affiliation(s)
- Jeremy D Coplan
- Division of Neuropsychopharmacology, Department of Psychiatry and Behavioral Science, State University of New York Downstate Medical Center , Brooklyn, NY , USA
| | - Srinath Gopinath
- Division of Neuropsychopharmacology, Department of Psychiatry and Behavioral Science, State University of New York Downstate Medical Center , Brooklyn, NY , USA
| | - Chadi G Abdallah
- Department of Psychiatry, Yale School of Medicine , New Haven, CT , USA ; Clinical Neuroscience Division, National Center for PTSD , West Haven, CT , USA
| | - Benjamin R Berry
- State University of New York Downstate College of Medicine , Brooklyn, NY , USA
| |
Collapse
|
20
|
Miyake A, Kitamura Y, Miyazaki I, Asanuma M, Sendo T. Effects of (+)-8-OH-DPAT on the duration of immobility during the forced swim test and hippocampal cell proliferation in ACTH-treated rats. Pharmacol Biochem Behav 2014; 122:240-5. [PMID: 24732636 DOI: 10.1016/j.pbb.2014.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 03/10/2014] [Accepted: 04/05/2014] [Indexed: 12/31/2022]
Abstract
In the present study, we examined the effect of ACTH on the immobilization of rats in the forced swim test and hippocampal cell proliferation after administration of the 5-HT1A receptor agonist, R-(+)-8-hydroxy-2-di-n-propylamino tetralin ((+)-8-OH-DPAT). Chronic treatment with (+)-8-OH-DPAT (0.01-0.1 mg/kg, s.c.) significantly decreased the duration of immobility in saline- and ACTH-treated rats. Chronic administration of ACTH caused a significant decrease in hippocampal cell proliferation. However, (+)-8-OH-DPAT significantly normalized cell proliferation in ACTH-treated rats. We then investigated the effects of (+)-8-OH-DPAT on the expression of brain-derived neurotrophic factor (BDNF) and cyclin D1 (elements of cyclic adenosine monophosphate response element-binding protein (CREB)-BDNF and Wnt signaling pathways, respectively) in the hippocampus of saline- and ACTH-treated rats. ACTH treatment significantly decreased the expression of cyclin D1, while treatment with (+)-8-OH-DPAT normalized the expression of cyclin D1 in ACTH-treated rats. However, the expression of BDNF did not change in either saline- or ACTH-treated rats. These findings suggest that the antidepressant effects of (+)-8-OH-DPAT in treatment-resistant animals may be attributed to an enhancement of hippocampal cell proliferation, at least in part due to an enhancement of cyclin D1 expression.
Collapse
Affiliation(s)
- Ayaka Miyake
- Department of Clinical Pharmacy, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Yoshihisa Kitamura
- Department of Clinical Pharmacy, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Ikuko Miyazaki
- Department of Brain Science, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Masato Asanuma
- Department of Brain Science, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Toshiaki Sendo
- Department of Clinical Pharmacy, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| |
Collapse
|
21
|
Improving cognition in schizophrenia with antipsychotics that elicit neurogenesis through 5-HT1A receptor activation. Neurobiol Learn Mem 2014; 110:72-80. [DOI: 10.1016/j.nlm.2013.12.015] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 12/21/2013] [Accepted: 12/27/2013] [Indexed: 01/20/2023]
|
22
|
Faure C, Mnie-Filali O, Haddjeri N. Long-term adaptive changes induced by serotonergic antidepressant drugs. Expert Rev Neurother 2014; 6:235-45. [PMID: 16466303 DOI: 10.1586/14737175.6.2.235] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The development of conventional antidepressants has been largely based on the hypothesis of monoaminergic dysfunctions and focuses particularly on the serotonin 5-hydroxytryptamine (5-HT) system. Hence, various classes of antidepressant treatments enhance 5-HT neurotransmission with a time course consistent with their delayed therapeutic effect. This delayed onset appears to be associated with the gradual development of specific adaptive changes of functional 5-HT receptors. However, recent theories suggest that major depressive disorders may be associated with impairments of functional plasticity and cellular flexibility. This review discusses several physiological mechanisms by which 5-HT function and hippocampal neuroplasticity are regulated. Knowledge of these long-term adaptations will increase not only our understanding of pathological processes underlying affective disorders, but could also lead to the development of new strategies to treat these devastating illnesses.
Collapse
Affiliation(s)
- Céline Faure
- Laboratoire de Neuropharmacologie et Neurochimie, Faculté de Pharmacie, Université Claude Bernard, Lyon 1, EA-512, 8, Avenue Rockefeller, 69373 Lyon Cedex 08, France.
| | | | | |
Collapse
|
23
|
Berg DA, Belnoue L, Song H, Simon A. Neurotransmitter-mediated control of neurogenesis in the adult vertebrate brain. Development 2013; 140:2548-61. [PMID: 23715548 DOI: 10.1242/dev.088005] [Citation(s) in RCA: 168] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It was long thought that no new neurons are added to the adult brain. Similarly, neurotransmitter signaling was primarily associated with communication between differentiated neurons. Both of these ideas have been challenged, and a crosstalk between neurogenesis and neurotransmitter signaling is beginning to emerge. In this Review, we discuss neurotransmitter signaling as it functions at the intersection of stem cell research and regenerative medicine, exploring how it may regulate the formation of new functional neurons and outlining interactions with other signaling pathways. We consider evolutionary and cross-species comparative aspects, and integrate available results in the context of normal physiological versus pathological conditions. We also discuss the potential role of neurotransmitters in brain size regulation and implications for cell replacement therapies.
Collapse
Affiliation(s)
- Daniel A Berg
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | | | | | |
Collapse
|
24
|
Diaz SL, Narboux-Nême N, Trowbridge S, Scotto-Lomassese S, Kleine Borgmann FB, Jessberger S, Giros B, Maroteaux L, Deneris E, Gaspar P. Paradoxical increase in survival of newborn neurons in the dentate gyrus of mice with constitutive depletion of serotonin. Eur J Neurosci 2013; 38:2650-8. [DOI: 10.1111/ejn.12297] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 06/06/2013] [Accepted: 06/06/2013] [Indexed: 12/21/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | - Evan Deneris
- Case Western Reserve University; Cleveland; OH; USA
| | | |
Collapse
|
25
|
Neural plasticity and proliferation in the generation of antidepressant effects: hippocampal implication. Neural Plast 2013; 2013:537265. [PMID: 23862076 PMCID: PMC3703717 DOI: 10.1155/2013/537265] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 05/01/2013] [Accepted: 05/08/2013] [Indexed: 12/15/2022] Open
Abstract
It is widely accepted that changes underlying depression and antidepressant-like effects involve not only alterations in the levels of neurotransmitters as monoamines and their receptors in the brain, but also structural and functional changes far beyond. During the last two decades, emerging theories are providing new explanations about the neurobiology of depression and the mechanism of action of antidepressant strategies based on cellular changes at the CNS level. The neurotrophic/plasticity hypothesis of depression, proposed more than a decade ago, is now supported by multiple basic and clinical studies focused on the role of intracellular-signalling cascades that govern neural proliferation and plasticity. Herein, we review the state-of-the-art of the changes in these signalling pathways which appear to underlie both depressive disorders and antidepressant actions. We will especially focus on the hippocampal cellularity and plasticity modulation by serotonin, trophic factors as brain-derived neurotrophic factor (BDNF), and vascular endothelial growth factor (VEGF) through intracellular signalling pathways—cAMP, Wnt/β-catenin, and mTOR. Connecting the classic monoaminergic hypothesis with proliferation/neuroplasticity-related evidence is an appealing and comprehensive attempt for improving our knowledge about the neurobiological events leading to depression and associated to antidepressant therapies.
Collapse
|
26
|
Saenz del Burgo L, Cortés R, Mengod G, Montaña M, García del Caño G, Sallés J. Chronic effects of corticosterone on GIRK1-3 subunits and 5-HT1A receptor expression in rat brain and their reversal by concurrent fluoxetine treatment. Eur Neuropsychopharmacol 2013; 23:229-39. [PMID: 22591911 DOI: 10.1016/j.euroneuro.2012.04.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 03/09/2012] [Accepted: 04/14/2012] [Indexed: 11/17/2022]
Abstract
Dysregulation of the serotonergic system and abnormalities of the hypothalamic-pituitary-adrenal axis have been demonstrated in major depression. Animal studies indicate that 5-HT1A receptor expression may be reduced by long-term administration of corticosterone. However, similar studies on the regulation of GIRK channels, one of the most important effectors of the neuronal 5-HT1A receptor, are limited. In order to address these issues, slow-release corticosterone pellets were implanted subcutaneously to adrenal intact male rats (200mg pellets, 35 days release). Starting on day 15, animals were treated for 21 days with fluoxetine (5mg/kg/day, i.p.), or vehicle. Using in situ hybridization histochemistry and receptor autoradiography, we found that chronic corticosterone treatment was accompanied by a significant decrease on the mRNAs coding for mineralocorticoid receptors in hippocampal areas. Under these conditions, 5-HT1A receptor mRNA expression decreased in dorsal raphe nucleus and dentate gyrus. However, 5-HT1A receptor levels, as measured by [(3)H]-8-OH-DPAT binding, diminished significantly only in dentate gyrus. It is noteworthy that chronic treatment with fluoxetine reversed the alterations on 5-HT1A receptor mRNA levels only in dorsal raphe. Finally, chronic corticosterone treatment produced an increase on the mRNA coding for the GIRK2 subunit in several hypothalamic and thalamic areas, which was reversed by fluoxetine. Measurements of cell density and volume of the granular layer of the dentate gyrus did not reveal significant changes after corticosterone or corticosterone plus fluoxetine treatments. These data are relevant for a better understanding of the differential regulation of pre- and postsynaptic 5-HT1A receptors by corticosterone flattened rhythm.
Collapse
MESH Headings
- Animals
- Autoradiography
- Corticosterone/pharmacology
- Dentate Gyrus/drug effects
- Dentate Gyrus/metabolism
- Depressive Disorder, Major/metabolism
- Fluoxetine/pharmacology
- G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects
- G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics
- G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism
- Gene Expression/drug effects
- Gene Expression Profiling
- Hippocampus/drug effects
- Hippocampus/metabolism
- Hypothalamo-Hypophyseal System/metabolism
- Male
- Pituitary-Adrenal System/metabolism
- Raphe Nuclei/drug effects
- Raphe Nuclei/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, Serotonin, 5-HT1A/drug effects
- Receptor, Serotonin, 5-HT1A/genetics
- Receptor, Serotonin, 5-HT1A/metabolism
- Receptors, Glucocorticoid/drug effects
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/metabolism
- Receptors, Mineralocorticoid/drug effects
- Receptors, Mineralocorticoid/genetics
- Receptors, Mineralocorticoid/metabolism
- Selective Serotonin Reuptake Inhibitors/pharmacology
- Thalamus/drug effects
- Thalamus/metabolism
Collapse
Affiliation(s)
- Laura Saenz del Burgo
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | | | | | | | | | | |
Collapse
|
27
|
Arnold SA, Hagg T. Serotonin 1A receptor agonist increases species- and region-selective adult CNS proliferation, but not through CNTF. Neuropharmacology 2012; 63:1238-47. [PMID: 22884499 PMCID: PMC3438376 DOI: 10.1016/j.neuropharm.2012.07.047] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 07/13/2012] [Accepted: 07/27/2012] [Indexed: 01/06/2023]
Abstract
Endogenous ciliary neurotrophic factor (CNTF)(1) regulates neurogenesis of the adult brain in the hippocampal subgranular zone (SGZ)(2) and the subventricular zone (SVZ)(3). We have previously shown that the cAMP-inhibiting D2 dopamine receptor increases neurogenesis by inducing astroglial CNTF expression. Here, we investigated the potential role of CNTF in the proliferative response to pharmacological stimulation of the serotonin 1A (5-HT1A)(4) receptor, which also inhibits cAMP, in adult mice and rats. Like others, we show that systemic treatment with the active R-enantiomer of the 5-HT1A agonist 8-Hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT)(5) induces proliferation in the SGZ in rats using unbiased stereology of 5-Bromo-2'-deoxyuridine (BrdU)(6) positive nuclei. However, despite the bioactivity of R-8-OH-DPAT, as also shown by a decrease in hippocampal nNOS(7) mRNA levels, it did not increase CNTF mRNA as shown by highly specific quantitative RT-PCR (qPCR)(8). Surprisingly, R-8-OH-DPAT did not cause an increase in SVZ proliferation in rats or in either the SVZ or SGZ of two different strains of mice, C57BL/6J, and 129SvEv, using acute or chronic treatments. There also were no changes in CNTF mRNA, and also not in mice treated with a widely used racemic mixture of 8-OH-DPAT, higher doses or after intracerebral injection, which reduced nNOS. In contrast to the others, we propose that the 5-HT1A receptor might be non-functional in mice with regards to regulating normal neurogenesis and has region-selective activities in rats. These species- and region-specific actions raise important questions about the role of the 5-HT1A receptor in human neurogenesis and its implications for the field of depression.
Collapse
Affiliation(s)
- Sheila A. Arnold
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Kentucky 40292
- Departments of Neurological Surgery, University of Louisville, Kentucky 40292
| | - Theo Hagg
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Kentucky 40292
- Departments of Neurological Surgery, University of Louisville, Kentucky 40292
- Pharmacology and Toxicology, University of Louisville, Kentucky 40292
| |
Collapse
|
28
|
Abstract
The importance of adult neurogenesis has only recently been accepted, resulting in a completely new field of investigation within stem cell biology. The regulation and functional significance of adult neurogenesis is currently an area of highly active research. G-protein-coupled receptors (GPCRs) have emerged as potential modulators of adult neurogenesis. GPCRs represent a class of proteins with significant clinical importance, because approximately 30% of all modern therapeutic treatments target these receptors. GPCRs bind to a large class of neurotransmitters and neuromodulators such as norepinephrine, dopamine, and serotonin. Besides their typical role in cellular communication, GPCRs are expressed on adult neural stem cells and their progenitors that relay specific signals to regulate the neurogenic process. This review summarizes the field of adult neurogenesis and its methods and specifies the roles of various GPCRs and their signal transduction pathways that are involved in the regulation of adult neural stem cells and their progenitors. Current evidence supporting adult neurogenesis as a model for self-repair in neuropathologic conditions, adult neural stem cell therapeutic strategies, and potential avenues for GPCR-based therapeutics are also discussed.
Collapse
Affiliation(s)
- Van A Doze
- Department of Molecular Cardiology, NB50, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | | |
Collapse
|
29
|
Masi G, Brovedani P. The hippocampus, neurotrophic factors and depression: possible implications for the pharmacotherapy of depression. CNS Drugs 2011; 25:913-31. [PMID: 22054117 DOI: 10.2165/11595900-000000000-00000] [Citation(s) in RCA: 186] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Depression is a prevalent, highly debilitating mental disorder affecting up to 15% of the population at least once in their lifetime, with huge costs for society. Neurobiological mechanisms of depression are still not well known, although there is consensus about interplay between genetic and environmental factors. Antidepressant medications are frequently used in depression, but at least 50% of patients are poor responders, even to more recently discovered medications. Furthermore, clinical response only occurs following weeks to months of treatment and only chronic treatment is effective, suggesting that actions beyond the rapidly occurring effect of enhancing monoaminergic systems, such as adaptation of these systems, are responsible for the effects of antidepressants. Recent studies indicate that an impairment of synaptic plasticity (neurogenesis, axon branching, dendritogenesis and synaptogenesis) in specific areas of the CNS, particularly the hippocampus, may be a core factor in the pathophysiology of depression. The abnormal neural plasticity may be related to alterations in the levels of neurotrophic factors, namely brain-derived neurotrophic factor (BDNF), which play a central role in plasticity. As BDNF is repressed by stress, epigenetic regulation of the BDNF gene may play an important role in depression. The hippocampus is smaller in depressed patients, although it is unclear whether smaller size is a consequence of depression or a pre-existing, vulnerability marker for depression. Environmental stressors triggering activation of the hypothalamic-pituitary-adrenal axis cause the brain to be exposed to corticosteroids, affecting neurobehavioural functions with a strong downregulation of hippocampal neurogenesis, and are a major risk factor for depression. Antidepressant treatment increases BDNF levels, stimulates neurogenesis and reverses the inhibitory effects of stress, but this effect is evident only after 3-4 weeks of administration, the time course for maturation of new neurons. The ablation of hippocampal neurogenesis blocks the behavioural effects of antidepressants in animal models. The above findings suggest new possible targets for the pharmacotherapy of depression such as neurotrophic factors, their receptors and related intracellular signalling cascades; agents counteracting the effects of stress on hippocampal neurogenesis (including antagonists of corticosteroids, inflammatory cytokines and their receptors); and agents facilitating the activation of gene expression and increasing the transcription of neurotrophins in the brain.
Collapse
Affiliation(s)
- Gabriele Masi
- IRCCS Stella Maris, Scientific Institute of Child Neurology and Psychiatry, Calambrone, Pisa, Italy.
| | | |
Collapse
|
30
|
Lu KT, Sun CL, Wo PYY, Yen HH, Tang TH, Ng MC, Huang ML, Yang YL. Hippocampal neurogenesis after traumatic brain injury is mediated by vascular endothelial growth factor receptor-2 and the Raf/MEK/ERK cascade. J Neurotrauma 2011; 28:441-50. [PMID: 21091268 DOI: 10.1089/neu.2010.1473] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Adult neurogenesis occurs in the subgranular zone of the hippocampal dentate gyrus, and can be modulated by physiological and pathological events. We examined the effect of vascular endothelial growth factor (VEGF), and the correlation between VEGF and the Raf/MEK/ERK cascade in neurogenesis after traumatic brain injury (TBI). The expression of VEGF and the phosphorylation level of Raf/MEK/ERK were analyzed by Western blot, and TBI-induced neurogenesis was determined by immunofluorescence labeling and confocal microscopic detection. Hippocampal VEGF began to increase after 12 h, and reached a peak at day 7. Along with the upregulation of VEGF, neurogenesis in the hippocampus also increased. Administration of the VEGF antisense oligodeoxynucleotide, or the VEGF receptor-2 antagonist SU1498 (10 μg, ICV), attenuated the phosphorylation of the MAPK cascade proteins and caused a decrease in neurogenesis in the hippocampus. Similarly, administration of the ERK inhibitor PD98059 (500 ng, ICV) also exhibited a suppressive effect on neurogenesis. Our results indicate that VEGF plays an important role in neurogenesis after TBI, and that the process involves VEGF receptor-2 and the Raf/MEK/ERK cascade.
Collapse
Affiliation(s)
- Kwok-Tung Lu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Pinnock SB, Blake AM, Platt NJ, Herbert J. The roles of BDNF, pCREB and Wnt3a in the latent period preceding activation of progenitor cell mitosis in the adult dentate gyrus by fluoxetine. PLoS One 2010; 5:e13652. [PMID: 21048974 PMCID: PMC2965105 DOI: 10.1371/journal.pone.0013652] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Accepted: 10/04/2010] [Indexed: 02/04/2023] Open
Abstract
The formation of new neurons continues into adult life in the dentate gyrus of the rat hippocampus, as in many other species. Neurogenesis itself turns out to be highly labile, and is regulated by a number of factors. One of these is the serotoninergic system: treatment with drugs (such as the SSRI fluoxetine) markedly stimulates mitosis in the progenitor cells of the dentate gyrus. But this process has one remarkable feature: it takes at least 14 days of continuous treatment to be effective. This is despite the fact that the pharmacological action of fluoxetine occurs within an hour or so of first administration. This paper explores the role of BDNF in this process, using the effect of a Trk antagonist (K252a) on the labelling of progenitor cells with the mitosis marker Ki67 and the associated expression of pCREB and Wnt3a. These experiments show that (i) Fluoxetine increased Ki67 counts, as well as pCREB and Wnt3a expression in the dentate gyrus. The action of fluoxetine on the progenitor cells and on pCREB (but not Wnt3a) depends upon Trk receptor activation, since it was prevented by icv infusion of K252a. (ii) These receptors are required for both the first 7 days of fluoxetine action, during which no apparent change in progenitor mitosis occurs, as well as the second 7 days. Increased pCREB was always associated with progenitor cell mitosis, but Wnt3a expression may be necessary but not sufficient for increased progenitor cell proliferation. These results shed new light on the action of fluoxetine on neurogenesis in the adult dentate gyrus, and have both clinical and experimental interest.
Collapse
Affiliation(s)
- Scarlett B Pinnock
- Department of Physiology, Cambridge Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
32
|
Polter AM, Li X. 5-HT1A receptor-regulated signal transduction pathways in brain. Cell Signal 2010; 22:1406-12. [PMID: 20363322 PMCID: PMC2903656 DOI: 10.1016/j.cellsig.2010.03.019] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Accepted: 03/25/2010] [Indexed: 10/19/2022]
Abstract
Serotonin is an influential monoamine neurotransmitter that signals through a number of receptors to modulate brain function. Among different serotonin receptors, the serotonin 1A (5-HT1A) receptors have been tied to a variety of physiological and pathological processes, notably in anxiety, mood, and cognition. 5-HT1A receptors couple not only to the classical inhibitory G protein-regulated signaling pathway, but also to signaling pathways traditionally regulated by growth factors. Despite the importance of 5-HT1A receptors in brain function, little is known about how these signaling mechanisms link 5-HT1A receptors to regulation of brain physiology and behavior. Following a brief summary of the known physiological and behavioral effects of 5-HT1A receptors, this article will review the signaling pathways regulated by 5-HT1A receptors, and discuss the potential implication of these signaling pathways in 5-HT1A receptor-regulated physiological processes and behaviors.
Collapse
Affiliation(s)
- Abigail M. Polter
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Xiaohua Li
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
33
|
Barr JL, Renner KJ, Forster GL. Withdrawal from chronic amphetamine produces persistent anxiety-like behavior but temporally-limited reductions in monoamines and neurogenesis in the adult rat dentate gyrus. Neuropharmacology 2010; 59:395-405. [PMID: 20638943 DOI: 10.1016/j.neuropharm.2010.05.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 05/24/2010] [Accepted: 05/25/2010] [Indexed: 11/28/2022]
Abstract
Acute amphetamine administration activates monoaminergic pathways and increases systemic corticosterone, both of which influence anxiety states and adult dentate gyrus neurogenesis. Chronic amphetamine increases anxiety states in rats when measured at 24 h and at 2 weeks of withdrawal. However, the effects of chronic amphetamine exposure and withdrawal on long term anxiety-like behavior and adult neurogenesis in the dentate gyrus are unknown. Adult male rats were administered amphetamine (2.5 mg/kg, ip.) daily for two weeks. Anxiety-like behaviors were increased markedly in amphetamine-treated rats following four weeks of withdrawal from amphetamine. Plasma corticosterone level was unaltered by amphetamine treatment or withdrawal. However, norepinephrine and serotonin concentrations were selectively reduced in the dentate gyrus 20 h following amphetamine treatment. This effect did not persist through the four week withdrawal period. In separate experiments, rats received bromodeoxyuridine to label cells in S-phase, prior to or immediately following amphetamine treatment. Newly generated cells were quantified to measure extent of progenitor cell proliferation and neurogenesis following treatment or withdrawal. Progenitor cell proliferation and neurogenesis were not significantly affected by amphetamine exposure when measured 20 h following the last amphetamine treatment. However, neurogenesis in the dentate gyrus was reduced after four weeks of withdrawal when compared to saline-pretreated rats. Overall, our findings indicate that withdrawal from chronic amphetamine leads to persistent anxiety-like behavior which may be maintained by reduced neurogenesis in the dentate gyrus at this protracted withdrawal time point. However, neurogenesis is unaffected at earlier withdrawal time points where anxiety states emerge, suggesting different mechanisms may underlie the emergence of anxiety states during amphetamine withdrawal.
Collapse
Affiliation(s)
- Jeffrey L Barr
- Division of Basic Biomedical Sciences and Neuroscience Group, Sanford School of Medicine, University of South Dakota, 414 E. Clark St., Vermillion, SD, USA
| | | | | |
Collapse
|
34
|
Soumier A, Banasr M, Kerkerian-Le Goff L, Daszuta A. Region- and phase-dependent effects of 5-HT(1A) and 5-HT(2C) receptor activation on adult neurogenesis. Eur Neuropsychopharmacol 2010; 20:336-45. [PMID: 20022222 DOI: 10.1016/j.euroneuro.2009.11.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Revised: 10/26/2009] [Accepted: 11/18/2009] [Indexed: 11/28/2022]
Abstract
Adult neurogenesis and serotoninergic transmission are associated to mood disorders and their treatments. The present study focused on the effects of chronic activation of 5-HT(1A) and 5-HT(2C) receptors on newborn cell survival in the dentate gyrus (DG) and olfactory bulb (OB), and examined whether potential neurogenic zones as the prefrontal cortex (PFC) and striatum (ST) are reactive to these treatments. Administration of 8-OH-DPAT, but not RO600,175 increases neurogenesis and survival of late differentiating cells (15-21days) in the DG. Both 8-OH-DPAT and RO600,175 increase neurogenesis in the OB, but only 8-OH-DPAT affected cell survival, inducing a parallel decrease in the number of BrdU cells in the OB and increase in the SVZ, which suggests an impaired migration. In the PFC and ST, 8-OH-DPAT and R0600,175 increase gliogenesis (NG2-labeled cells). This study provides new insights on the serotonergic regulation of critical phases of neurogenesis helpful to understand the neurogenic and gliogenic effects of antidepressant treatments in different brain regions.
Collapse
Affiliation(s)
- Amélie Soumier
- IBDML, UMR, CNRS-Université de la Mediterranée, Marseille, France
| | | | | | | |
Collapse
|
35
|
Tharayil VS, Wouters MM, Stanich JE, Roeder JL, Lei S, Beyder A, Gomez-Pinilla PJ, Gershon MD, Maroteaux L, Gibbons SJ, Farrugia G. Lack of serotonin 5-HT2B receptor alters proliferation and network volume of interstitial cells of Cajal in vivo. Neurogastroenterol Motil 2010; 22:462-9, e109-10. [PMID: 19941613 PMCID: PMC2852486 DOI: 10.1111/j.1365-2982.2009.01435.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Normal gastrointestinal motility requires intact networks of interstitial cells of Cajal (ICC). Interstitial cells of Cajal numbers are maintained by a balance between cell loss factors and survival/trophic/growth factors. Activation of 5-HT(2B) receptors expressed on ICC increases ICC proliferation in vitro. It is not known whether 5-HT(2B) receptors on ICC are activated in vivo. The aims of this study were to investigate if adult ICC proliferate, whether the proliferation of ICC in vivo is affected by knocking out the 5-HT(2B) receptor, and if alterations in proliferation affect ICC networks. METHODS Proliferating ICC were identified by immunoreactivity for Ki67 in both the myenteric and deep muscular plexus regions of the jejunum in mice with a targeted insertion of a neomycin resistance cassette into the second coding exon of the htr2b receptor gene. KEY RESULTS Adult ICC do proliferate. The number of proliferating ICC was lower in the myenteric plexus region of Htr2b(-/-) compared to Htr2b(+/+) mice. The volume of Kit-positive ICC was 30% lower in the myenteric plexus region and 40% lower in the deep muscular plexus region in Htr2b(-/-) mice where the number of ICC was also reduced. CONCLUSIONS & INFERENCES Interstitial cells of Cajal proliferate in adult mice and activation of 5-HT(2B) receptors results in increased proliferation of ICC in vivo. Furthermore, lack of 5-HT(2B) receptor signaling reduces the density of ICC networks in mature mice. These data suggest that 5-HT(2B) receptor signaling is required for maintenance of ICC networks, adding 5-HT to the growing number of factors shown to regulate ICC networks.
Collapse
Affiliation(s)
- Vivek S. Tharayil
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases, Mayo Clinic, Rochester, MN, USA
| | - Mira M. Wouters
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases, Mayo Clinic, Rochester, MN, USA
| | - Jennifer E. Stanich
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases, Mayo Clinic, Rochester, MN, USA
| | - Jaime L. Roeder
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases, Mayo Clinic, Rochester, MN, USA
| | - Sha Lei
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases, Mayo Clinic, Rochester, MN, USA
| | - Arthur Beyder
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases, Mayo Clinic, Rochester, MN, USA
| | - Pedro J. Gomez-Pinilla
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases, Mayo Clinic, Rochester, MN, USA
| | - Michael D. Gershon
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Luc Maroteaux
- INSERM, U839, Institut du Fer à Moulin, 17 rue du Fer à Moulin, 75005 Paris, France
| | - Simon J. Gibbons
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases, Mayo Clinic, Rochester, MN, USA, Dept of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Gianrico Farrugia
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases, Mayo Clinic, Rochester, MN, USA, Dept of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
36
|
Benninghoff J, Gritti A, Rizzi M, Lamorte G, Schloesser RJ, Schmitt A, Robel S, Genius J, Moessner R, Riederer P, Manji HK, Grunze H, Rujescu D, Moeller HJ, Lesch KP, Vescovi AL. Serotonin depletion hampers survival and proliferation in neurospheres derived from adult neural stem cells. Neuropsychopharmacology 2010; 35:893-903. [PMID: 20010549 PMCID: PMC3055363 DOI: 10.1038/npp.2009.181] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Revised: 09/04/2009] [Accepted: 09/07/2009] [Indexed: 02/08/2023]
Abstract
Serotonin (5-HT) and the serotonergic system have recently been indicated as modulators of adult hippocampal neurogenesis. In this study, we evaluated the role of 5-HT on the functional features in neurospheres derived from adult neural stem cells (ANSC). We cultured neurospheres derived from mouse hippocampus in serum-free medium containing epidermal (EGF) and type-2 fibroblast growth factor (FGF2). Under these conditions ANSC expressed both isoforms of tryptophane-hydroxylase (TPH) and produced 5-HT. Blocking TPH function by para-chlorophenylalanine (PCPA) reduced ANSC proliferation, which was rescued by exogenous 5-HT. 5-HT action on ANSC was mediated predominantly by the serotonin receptor subtype 5-HT1A and, to a lesser extent, through the 5-HT2C (receptor) subtype, as shown by selectively antagonizing these receptors. Finally, we documented a 5-HT-induced increase of ANSC migration activity. In summary, we demonstrated a powerful serotonergic impact on ANSC functional features, which was mainly mediated by 5-HT1A receptors.
Collapse
Affiliation(s)
- Jens Benninghoff
- S. Raffaele Scientific Institute, Stem Cell Research Institute (HSR-SCRI), Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Castro JE, Varea E, Márquez C, Cordero MI, Poirier G, Sandi C. Role of the amygdala in antidepressant effects on hippocampal cell proliferation and survival and on depression-like behavior in the rat. PLoS One 2010; 5:e8618. [PMID: 20062812 PMCID: PMC2799663 DOI: 10.1371/journal.pone.0008618] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Accepted: 12/13/2009] [Indexed: 11/19/2022] Open
Abstract
The stimulation of adult hippocampal neurogenesis by antidepressants has been associated with multiple molecular pathways, but the potential influence exerted by other brain areas has received much less attention. The basolateral complex of the amygdala (BLA), a region involved in anxiety and a site of action of antidepressants, has been implicated in both basal and stress-induced changes in neural plasticity in the dentate gyrus. We investigated here whether the BLA modulates the effects of the SSRI antidepressant fluoxetine on hippocampal cell proliferation and survival in relation to a behavioral index of depression-like behavior (forced swim test). We used a lesion approach targeting the BLA along with a chronic treatment with fluoxetine, and monitored basal anxiety levels given the important role of this behavioral trait in the progress of depression. Chronic fluoxetine treatment had a positive effect on hippocampal cell survival only when the BLA was lesioned. Anxiety was related to hippocampal cell survival in opposite ways in sham- and BLA-lesioned animals (i.e., negatively in sham- and positively in BLA-lesioned animals). Both BLA lesions and low anxiety were critical factors to enable a negative relationship between cell proliferation and depression-like behavior. Therefore, our study highlights a role for the amygdala on fluoxetine-stimulated cell survival and on the establishment of a link between cell proliferation and depression-like behavior. It also reveals an important modulatory role for anxiety on cell proliferation involving both BLA-dependent and -independent mechanisms. Our findings underscore the amygdala as a potential target to modulate antidepressants' action in hippocampal neurogenesis and in their link to depression-like behaviors.
Collapse
Affiliation(s)
- Jorge E. Castro
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Emilio Varea
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Cristina Márquez
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Maria Isabel Cordero
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Guillaume Poirier
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
38
|
Wouters MM, Roeder JL, Tharayil VS, Stanich JE, Strege PR, Lei S, Bardsley MR, Ordog T, Gibbons SJ, Farrugia G. Protein kinase C{gamma} mediates regulation of proliferation by the serotonin 5-hydroxytryptamine receptor 2B. J Biol Chem 2009; 284:21177-84. [PMID: 19531484 DOI: 10.1074/jbc.m109.015859] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of the 5-hydroxytryptamine receptor 2B (5-HT(2B)), a G(q/11) protein-coupled receptor, results in proliferation of various cell types. The 5-HT(2B) receptor is also expressed on the pacemaker cells of the gastrointestinal tract, the interstitial cells of Cajal (ICC), where activation triggers ICC proliferation. The goal of this study was to characterize the mitogenic signal transduction cascade activated by the 5-HT(2B) receptor. All of the experiments were performed on mouse small intestine primary cell cultures. Activation of the 5-HT(2B) receptor by its agonist BW723C86 induced proliferation of ICC. Inhibition of phosphatidylinositol 3-kinase by LY294002 decreased base-line proliferation but had no effect on 5-HT(2B) receptor-mediated proliferation. Proliferation of ICC through the 5-HT(2B) receptor was inhibited by the phospholipase C inhibitor U73122 and by the inositol 1,4,5-trisphosphate receptor inhibitor Xestospongin C. Calphostin C, the alpha, beta, gamma, and micro protein kinase C (PKC) inhibitor Gö6976, and the alpha, beta, gamma, delta, and zeta PKC inhibitor Gö6983 inhibited 5-HT(2B) receptor-mediated proliferation, indicating the involvement of PKC alpha, beta, or gamma. Of all the PKC isoforms blocked by Gö6976, PKCgamma and micro mRNAs were found by single-cell PCR to be expressed in ICC. 5-HT(2B) receptor activation in primary cell cultures obtained from PKCgamma(-/-) mice did not result in a proliferative response, further indicating the requirement for PKCgamma in the proliferative response to 5-HT(2B) receptor activation. The data demonstrate that the 5-HT(2B) receptor-induced proliferative response of ICC is through phospholipase C, [Ca(2+)](i), and PKCgamma, implicating this PKC isoform in the regulation of cellular proliferation.
Collapse
Affiliation(s)
- Mira M Wouters
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Miles and Shirley Fiterman Center for Digestive Diseases, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Savitz J, Lucki I, Drevets WC. 5-HT(1A) receptor function in major depressive disorder. Prog Neurobiol 2009; 88:17-31. [PMID: 19428959 DOI: 10.1016/j.pneurobio.2009.01.009] [Citation(s) in RCA: 418] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2008] [Revised: 12/22/2008] [Accepted: 01/29/2009] [Indexed: 10/21/2022]
Abstract
Dysfunction of the serotonin 1A receptor (5-HT(1A)) may play a role in the genesis of major depressive disorder (MDD). Here we review the pharmacological, post-mortem, positron emission tomography (PET), and genetic evidence in support of this statement. We also touch briefly on two MDD-associated phenotypes, cognitive impairment and somatic pain. The results of pharmacological challenge studies with 5-HT(1A) receptor agonists are indicative of blunted endocrine responses in depressed patients. Lithium, valproate, selective serotonin reuptake inhibitors (SSRIs), tricyclic antidepressants (TCAs), and other treatment, such as electroconvulsive shock therapy (ECT), all increase post-synaptic 5-HT(1A) receptor signaling through either direct or indirect effects. Reduced somatodendritic and postsynaptic 5-HT(1A) receptor numbers or affinity have been reported in some post-mortem studies of suicide victims, a result consistent with well-replicated PET analyses demonstrating reduced 5-HT(1A) receptor binding potential in diverse regions such as the dorsal raphe, medial prefrontal cortex (mPFC), amygdala and hippocampus. 5-HT(1A) receptor knockout (KO) mice display increased anxiety-related behavior, which, unlike in their wild-type counterparts, cannot be rescued with antidepressant drug (AD) treatment. In humans, the G allele of a single nucleotide polymorphism (SNP) in the 5-HT(1A) receptor gene (HTR1A; rs6295), which abrogates a transcription factor binding site for deformed epidermal autoregulatory factor-1 (Deaf-1) and Hes5, has been reported to be over-represented in MDD cases. Conversely, the C allele has been associated with better response to AD drugs. We raise the possibility that 5-HT(1A) receptor dysfunction represents one potential mechanism underpinning MDD and other stress-related disorders.
Collapse
Affiliation(s)
- Jonathan Savitz
- Section on Neuroimaging in Mood and Anxiety Disorders, Mood and Anxiety Disorders Program, NIH/NIMH, Bethesda, MD 20892, United States.
| | | | | |
Collapse
|
40
|
Goodyer IM. Emanuel Miller Lecture: early onset depressions--meanings, mechanisms and processes. J Child Psychol Psychiatry 2008; 49:1239-56. [PMID: 19120706 DOI: 10.1111/j.1469-7610.2008.01964.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Depressive syndromes in children and adolescents constitute a serious group of mental disorders with considerable risk for recurrence. A more precise understanding of aetiology is necessary to improve treatment and management. METHODS Three neuroactive agents are purported to be involved in the aetiology of these disorders: serotonin, brain-derived neurotrophic factor and cortisol. A literature review was conducted to determine their contributions to the emergence of unipolar depressions in the adolescent years. RESULTS Serotonin, brain-derived neurotrophic factor and cortisol may operate in concert within two distinct functional frameworks: atypical early epigenesis arising in the first few years of life and resulting in the formation of a vulnerable neuronal network involving in particular the amygdala and ventral prefrontal cortex. Individuals with this vulnerability are likely to show impaired mood regulation when faced with environmental demands during adolescence and over the subsequent decades; and acquired neuroendangerment, a pathological brain process leading to reduced synaptic plasticity, in particular in the hippocampus and perhaps the nucleus accumbens and ventral tegmentum. This may result in motivational, cognitive and behavioural deficits at any point in the lifespan most apparent at times of environmental demand. CONCLUSIONS The characteristics, course and outcome of a depressive episode may depend on the extent of the involvement of both atypical early neurogenesis and acquired neuroendangerment.
Collapse
Affiliation(s)
- Ian M Goodyer
- Developmental Psychiatry Section, Department of Psychiatry, University of Cambridge, UK.
| |
Collapse
|
41
|
Pinnock SB, Lazic SE, Wong HT, Wong IHW, Herbert J. Synergistic effects of dehydroepiandrosterone and fluoxetine on proliferation of progenitor cells in the dentate gyrus of the adult male rat. Neuroscience 2008; 158:1644-51. [PMID: 19068226 DOI: 10.1016/j.neuroscience.2008.10.035] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Revised: 10/06/2008] [Accepted: 10/15/2008] [Indexed: 11/24/2022]
Abstract
The 5-HT re-uptake inhibitor (SSRI) fluoxetine and the adrenal hormone dehydroepiandrosterone (DHEA) both increase the proliferation of progenitor cells in the adult hippocampus and also have antidepressant activity. This paper explores the combined ability of fluoxetine and DHEA to affect this process in the dentate gyrus of adult rats. We show that DHEA can render an otherwise ineffective dose of fluoxetine (2.5 mg/kg) able to increase progenitor cell proliferation to the same extent as doses four times higher (10 mg/kg). This synergistic action does not appear to be mediated by alterations in brain-derived neurotrophic factor (BDNF) gene expression; or by TrkB, mineralocorticoid, glucocorticoid, or 5-HT (5HT1A) receptor expression in the dentate gyrus; or by altered levels of plasma corticosterone. In a second experiment, the synergism between DHEA and fluoxetine was replicated. Furthermore, flattening the diurnal rhythm of plasma corticosterone by implanting additional corticosterone pellets s.c. prevented the effect of fluoxetine on progenitor cell division. This was not overcome by simultaneous treatment with DHEA, despite the latter's reported anti-glucocorticoid actions. The cellular mechanism for the potentiating action of DHEA on the pro- proliferative effects of fluoxetine in the adult hippocampus remains to be revealed. Since altered neurogenesis has been linked to the onset or recovery from depression, one consequence of these results is to suggest DHEA as a useful adjunct therapy for depression.
Collapse
Affiliation(s)
- S B Pinnock
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | | | | | | | | |
Collapse
|
42
|
Liew CJ, Lim YM, Bonwetsch R, Shamim S, Sato S, Reeves-Tyer P, Herscovitch P, Dustin I, Bagic A, Giovacchini G, Theodore WH. 18F-FCWAY and 18F-FDG PET in MRI-negative temporal lobe epilepsy. Epilepsia 2008; 50:234-9. [PMID: 18801033 DOI: 10.1111/j.1528-1167.2008.01789.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Positron emission tomography (PET) with (18)F-fluorodeoxyglucose (FDG) shows widespread hypometabolism even in temporal lobe epilepsy (TLE) patients with mesial temporal foci. (18)F-trans-4-fluoro-N-2-[4-(2-methoxyphenyl) piperazin-1-yl]ethyl-N-(2-pyridyl)cyclohexane carboxamide ((18)F-FCWAY) PET may show more specific 5-HT(1A)-receptor binding reduction in seizure initiation than in propagation regions. (18)FCWAY PET might be valuable for detecting epileptic foci, and distinguishing mesial from lateral temporal foci in MRI-negative patients with TLE. METHODS We performed (18)F-FCWAY-PET and (18)F-FDG-PET in 12 MRI-negative TLE patients who had had either surgery or subdural electrode recording, and 15 healthy volunteers. After partial volume correction for brain atrophy, free fraction-corrected volume of distribution (V/f1) measurement and asymmetry indices (AIs) were computed. We compared (18)F-FCWAY-PET and (18)F-FDG-PET results with scalp video electroencephalography (EEG), invasive EEG, and surgical outcome. RESULTS Mean (18)F-FCWAY V/f1, compared with normal controls, was decreased significantly in fusiform gyrus, hippocampus, and parahippocampus ipsilateral to epileptic foci, and AIs were significantly greater in hippocampus, parahippocampus, fusiform gyrus, amygdala, and inferior temporal regions. Eleven patients had clearly lateralized epileptogenic zones. Nine had congruent, and two nonlateralized, (18)F-FCWAY PET. One patient with bitemporal seizure onset had nonlateralized (18)F-FCWAY-PET. (18)F-FDG-PET showed congruent hypometabolism in 7 of 11 EEG-lateralized patients, bilateral hypometabolic regions in one, contralateral hypometabolism in one, as well as lateralized hypometabolism in the patient with bitemporal subdural seizure onset. Patients with mesial temporal foci tended to have lower superior and midtemporal (18)F-FCWAY V/f1 binding AI than those with lateral or diffuse foci. CONCLUSION (18)F-FCWAY-PET can detect reduced binding in patients with normal MRI, and may be more accurate than (18)F-FDG-PET.
Collapse
Affiliation(s)
- Clarissa J Liew
- Clinical Epilepsy Section, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Pinnock SB, Herbert J. Brain-derived neurotropic factor and neurogenesis in the adult rat dentate gyrus: interactions with corticosterone. Eur J Neurosci 2008; 27:2493-500. [PMID: 18547240 DOI: 10.1111/j.1460-9568.2008.06250.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Flattening the diurnal corticosterone rhythm prevented the stimulating action of L-NAME (a nitric oxide synthase, NOS, inhibitor) on progenitor cell proliferation in the dentate gyrus in Lister-Hooded adult male rats. The increased expression of brain-derived neurotrophic factor (BDNF) and trkB mRNA in the dentate gyrus which otherwise occurred after L-NAME was also prevented by clamping the corticoid rhythm in adrenalectomized rats, but was restored by daily additional injections of corticosterone (which replicates the diurnal rhythm). Unilateral infusions of BDNF into the lateral ventricle increased proliferation in the dentate gyrus on the side of the infusion, but this was not observed following implantation of subcutaneous corticosterone, which flattened the diurnal corticosterone rhythm. 5HT1A mRNA in the dentate gyrus was increased on both sides of the brain by unilateral BDNF infusions, but this was also prevented by subcutaneous corticosterone pellets. These results show that the diurnal rhythm of corticosterone regulates the stimulating action of NOS inhibitors on BDNF as well as on neurogenesis in the dentate gyrus, and that BDNF becomes ineffective on both proliferation rates and 5HT1A expression in the absence of a rhythm in corticosterone. This, together with our previous findings, suggests that corticoid rhythms permit both serotonin and NO access to BDNF, and the latter to regulate progenitor cell activity.
Collapse
Affiliation(s)
- Scarlett B Pinnock
- Department of Physiology, Development and Neuroscience, and Cambridge Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
44
|
Balu DT, Lucki I. Adult hippocampal neurogenesis: regulation, functional implications, and contribution to disease pathology. Neurosci Biobehav Rev 2008; 33:232-52. [PMID: 18786562 DOI: 10.1016/j.neubiorev.2008.08.007] [Citation(s) in RCA: 272] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 08/13/2008] [Accepted: 08/14/2008] [Indexed: 12/28/2022]
Abstract
It is now well established that the mammalian brain has the capacity to produce new neurons into adulthood. One such region that provides the proper milieu to sustain progenitor cells and is permissive to neuronal fate determination is located in the dentate gyrus of the hippocampus. This review will discuss in detail the complex process of adult hippocampal neurogenesis, including proliferation, differentiation, survival, and incorporation into neuronal networks. The regulation of this phenomenon by a number of factors is described, including neurotransmitter systems, growth factors, paracrine signaling molecules, neuropeptides, transcription factors, endogenous psychotropic systems, sex hormones, stress, and others. This review also addresses the functional significance of adult born hippocampal granule cells with regard to hippocampal circuitry dynamics and behavior. Furthermore, the relevance of perturbations in adult hippocampal neurogenesis to the pathophysiology of various disease states, including depression, schizophrenia, epilepsy, and diabetes are examined. Finally, this review discusses the potential of using hippocampal neurogenesis as a therapeutic target for these disorders.
Collapse
Affiliation(s)
- Darrick T Balu
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
45
|
Zetzsche T, Preuss UW, Bondy B, Frodl T, Zill P, Schmitt G, Koutsouleris N, Rujescu D, Born C, Reiser M, Möller HJ, Meisenzahl EM. 5-HT1A receptor gene C -1019 G polymorphism and amygdala volume in borderline personality disorder. GENES BRAIN AND BEHAVIOR 2008; 7:306-13. [PMID: 18387137 DOI: 10.1111/j.1601-183x.2007.00353.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Alterations of amygdala structure and function have been repeatedly described in patients with borderline personality disorder (BPD). The aim of our study was to determine whether a functional polymorphism of the 5-hydroxytryptamine(1A) receptor (5-HTR(1A)) gene C -1019 G (identity number: rs6295 G/C) is associated with structural changes of the amygdala in patients with BPD. Twenty-five right-handed female inpatients with BPD according to DSM IV and 25 healthy controls matched for age, sex, handedness and educational status were enrolled. Brain volumetry of the amygdala was performed with a 1.5-T Magnetom Vision apparatus (Siemens, Erlangen, Germany) and analyzed by the software program 'BRAINS'. Patients who have the 5-HTR(1A) gene G allele had significantly smaller amygdala volumes than C/C genotype carriers (P = 0.02). While no difference of allelic distribution between patients and controls was detected, the described effect of 5-HTR(1A) genotype on amygdala volume was found for the whole group of patients, as well as in the subgroup of patients with comorbid major depression (P = 0.004) but not in controls. In contrast to these subgroups of BPD patients who had significant amygdala volume differences, the mean amygdala volume of the whole group of BPD patients was not significantly different from that of controls. In summary, our study provides first evidence that 5-HTR(1A) gene C -1019 G polymorphism is associated with structural changes in the limbic system of BPD patients, a finding that might be disease related and might contribute to explanation of previous discrepant results regarding amygdala volume changes in BPD. Future research is recommended to clarify possible interactions between this functional polymorphism and symptoms, course and treatment responses in this disorder.
Collapse
Affiliation(s)
- T Zetzsche
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians University, Munich, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mueller AD, Pollock MS, Lieblich SE, Epp JR, Galea LAM, Mistlberger RE. Sleep deprivation can inhibit adult hippocampal neurogenesis independent of adrenal stress hormones. Am J Physiol Regul Integr Comp Physiol 2008; 294:R1693-703. [DOI: 10.1152/ajpregu.00858.2007] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Sleep deprivation (SD) can suppress cell proliferation in the hippocampal dentate gyrus of adult male rodents, suggesting that sleep may contribute to hippocampal functions by promoting neurogenesis. However, suppression of cell proliferation in rats by the platform-over-water SD method has been attributed to elevated corticosterone (Cort), a potent inhibitor of cell proliferation and nonspecific correlate of this procedure. We report here results that do not support this conclusion. Intact and adrenalectomized (ADX) male rats were subjected to a 96-h SD using multiple- and single-platform methods. New cells were identified by immunoreactivity for 5-bromo-2′-deoxyuridine (BrdU) or Ki67 and new neurons by immunoreactivity for BrdU and doublecortin. EEG recordings confirmed a 95% deprivation of rapid eye movement (REM) sleep and a 40% decrease of non-REM sleep. Cell proliferation in the dentate gyrus was suppressed by up to 50% in sleep-deprived rats relative to apparatus control or home cage control rats. This effect was also observed in ADX rats receiving continuous low-dose Cort replacement via subcutaneous minipumps but not in ADX rats receiving Cort replacement via drinking water. In these latter rats, Cort intake via water was reduced by 60% during SD; upregulation of cell proliferation by reduced Cort intake may obscure inhibitory effects of sleep loss on cell proliferation. SD had no effect on the percentage of new cells expressing a neuronal phenotype. These results demonstrate that the Cort replacement method is critical for detecting an effect of SD on cell proliferation and support a significant role for sleep in adult neurogenesis.
Collapse
|
47
|
Sørensen C, Øverli Ø, Summers CH, Nilsson GE. Social Regulation of Neurogenesis in Teleosts. BRAIN, BEHAVIOR AND EVOLUTION 2007; 70:239-46. [PMID: 17914255 DOI: 10.1159/000105487] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Salmonid fishes such as the rainbow trout (Oncorhynchus mykiss) are frequently used to study behavioral and neuroendocrine effects of socially induced stress. A predictable aggressive response to territorial intrusion, a well described neuroanatomy, and many essential similarities in the stress response in fishes and other vertebrates are among the advantages of this comparative model. One conspicuous difference when compared to mammals, however, is that in teleost fish and other non-mammalian vertebrates, neurogenesis persists into adulthood to a much higher degree. Very little is known about the functional significance of individual differences in the rate of brain cell proliferation in fish, or whether structural changes in the fish brain are influenced by the social environment. In this paper we discuss the observation that brain cell proliferation is reduced in subordinate fish, focusing in particular on whether such individual variation reflects a difference in coping style or is indeed a response to social interactions.
Collapse
|
48
|
Wouters MM, Gibbons SJ, Roeder JL, Distad M, Ou Y, Strege PR, Szurszewski JH, Farrugia G. Exogenous serotonin regulates proliferation of interstitial cells of Cajal in mouse jejunum through 5-HT2B receptors. Gastroenterology 2007; 133:897-906. [PMID: 17854596 DOI: 10.1053/j.gastro.2007.06.017] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2006] [Accepted: 05/31/2007] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Interstitial cells of Cajal (ICC) are required for normal gastrointestinal motility. Loss of ICC is associated with several motility disorders. The mechanisms modulating ICC survival and proliferation are poorly understood. This study aimed to establish whether 5-hydroxytryptamine (5-HT) plays a role in regulating ICC proliferation. METHODS Expression of 5-HT receptor mRNA was investigated in muscle strips, in purified populations of ICC, and in identified single cells. The effect of 5-HT(2B) receptor ligands on ICC numbers was studied in primary cell cultures. Proliferation of ICC was determined by counting Ki67-positive cells in culture. RESULTS Of the 5-HT receptors known to be involved in proliferation, 5-HT(2B) receptor mRNA was detected by reverse transcriptase-polymerase chain reaction (RT-PCR) in jejunal muscle, whereas 5-HT(1A), 5-HT(1D), and 5-HT(2C) receptor mRNAs were not. 5-HT(2B) receptor mRNA was found in single ICC and cells purified by flow cytometry. Exogenous 5-HT (1 micromol/L) increased (66% +/- 9%, P < .005) ICC numbers in culture. The 5-HT(2) receptor antagonist, ritanserin, and the 5-HT(2B) receptor antagonist, SB204741, inhibited the effect of 5-HT. The 5-HT(2B) receptor agonist BW 723C86 induced a concentration-dependent increase in ICC number (50% +/- 6% at 50 nM, P < .04) and increased ICC proliferation (25% +/- 3% vs 19 +/- 1% in controls, P < .03). CONCLUSIONS These studies establish that 5-HT(2B) receptors are expressed on ICC. Exogenous 5-HT regulates ICC numbers through 5-HT(2B) receptors in part by increasing ICC proliferation. The 5-HT(2B) receptor may serve as a novel pathway to regulate ICC numbers.
Collapse
MESH Headings
- Animals
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Cell Survival/physiology
- Coculture Techniques
- Indoles/pharmacology
- Jejunum/cytology
- Jejunum/drug effects
- Jejunum/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred Strains
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, Serotonin, 5-HT2B/genetics
- Receptor, Serotonin, 5-HT2B/metabolism
- Ritanserin/pharmacology
- Serotonin/pharmacology
- Serotonin 5-HT2 Receptor Agonists
- Serotonin Agents/pharmacology
- Serotonin Antagonists/pharmacology
- Thiophenes/pharmacology
- Urea/analogs & derivatives
- Urea/pharmacology
Collapse
Affiliation(s)
- Mira M Wouters
- Enteric Neuroscience Program, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Wouters MM, Farrugia G, Schemann M. 5-HT receptors on interstitial cells of Cajal, smooth muscle and enteric nerves. Neurogastroenterol Motil 2007; 19 Suppl 2:5-12. [PMID: 17620082 DOI: 10.1111/j.1365-2982.2007.00963.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The majority of the body's serotonin (5-HT) is produced by the gastrointestinal tract. 5-HT has several functions in the gastrointestinal tract. 5-HT is a paracrine signalling molecule released from enterochromaffin cells, a survival and proliferating factor and a neurotransmitter. The actions of 5-HT are transduced by a large family of 5-HT receptors, several of which are expressed on different gastrointestinal cell types including enteric nerves, smooth muscle and interstitial cells of Cajal (ICC). This review will summarize recent advances in understanding the role of 5-HT in regulating function of ICC, and the expression and function of 5-HT receptors on muscle and enteric nerves in human tissue. Rodent ICC express several 5-HT receptors including 5-HT(2B) receptors which regulate ICC survival and proliferation. Human smooth muscle and enteric neurons also express several 5-HT receptor subtypes. Expression and function of these receptors is significantly different from small laboratory animals. 5-HT(7) receptor activation causes relaxation of muscle, whereas 5-HT(2B) receptors increase muscle activity. The 5-HT(4) receptor appears to mediate both inhibition and activation of smooth muscle involving myogenic as well as neural actions. Despite the abundant expression of 5-HT(3) receptors in the human enteric nervous system no functional correlate has been as yet demonstrated.
Collapse
Affiliation(s)
- M M Wouters
- Enteric Neuroscience Program, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | |
Collapse
|
50
|
Joëls M, Karst H, Krugers HJ, Lucassen PJ. Chronic stress: implications for neuronal morphology, function and neurogenesis. Front Neuroendocrinol 2007; 28:72-96. [PMID: 17544065 DOI: 10.1016/j.yfrne.2007.04.001] [Citation(s) in RCA: 277] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2007] [Revised: 04/12/2007] [Accepted: 04/20/2007] [Indexed: 12/19/2022]
Abstract
In normal life, organisms are repeatedly exposed to brief periods of stress, most of which can be controlled and adequately dealt with. The presently available data indicate that such brief periods of stress have little influence on the shape of neurons or adult neurogenesis, yet change the physiological function of cells in two time-domains. Shortly after stress excitability in limbic areas is rapidly enhanced, but also in brainstem neurons which produce catecholamines; collectively, during this phase the stress hormones promote focused attention, alertness, vigilance and the initial steps in encoding of information linked to the event. Later on, when the hormone concentrations are back to their pre-stress level, gene-mediated actions by corticosteroids reverse and normalize the enhanced excitability, an adaptive response meant to curtail defense reactions against stressors and to enable further storage of relevant information. When stress is experienced repetitively in an uncontrollable and unpredictable manner, a cascade of processes in brain is started which eventually leads to profound, region-specific alterations in dendrite and spine morphology, to suppression of adult neurogenesis and to inappropriate functional responses to a brief stress exposure including a sensitized activation phase and inadequate normalization of brain activity. Although various compounds can effectively prevent these cellular changes by chronic stress, the exact mechanism by which the effects are accomplished is poorly understood. One of the challenges for future research is to link the cellular changes seen in animal models for chronic stress to behavioral effects and to understand the risks they can impose on humans for the precipitation of stress-related disorders.
Collapse
Affiliation(s)
- Marian Joëls
- SILS-CNS, University of Amsterdam, Kruislaan 320, 1098 SM Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|