51
|
Krol A, Wimmer RD, Halassa MM, Feng G. Thalamic Reticular Dysfunction as a Circuit Endophenotype in Neurodevelopmental Disorders. Neuron 2018; 98:282-295. [PMID: 29673480 PMCID: PMC6886707 DOI: 10.1016/j.neuron.2018.03.021] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 01/30/2018] [Accepted: 03/12/2018] [Indexed: 02/06/2023]
Abstract
Diagnoses of behavioral disorders such as autism spectrum disorder and schizophrenia are based on symptomatic descriptions that have been difficult to connect to mechanism. Although psychiatric genetics provide insight into the genetic underpinning of such disorders, with a majority of cases explained by polygenic factors, it remains difficult to design rational treatments. In this review, we highlight the value of understanding neural circuit function both as an intermediate level of explanatory description that links gene to behavior and as a pathway for developing rational diagnostics and therapeutics for behavioral disorders. As neural circuits perform hierarchically organized computational functions and give rise to network-level processes (e.g., macroscopic rhythms and goal-directed or homeostatic behaviors), correlated network-level deficits may indicate perturbation of a specific circuit. Therefore, identifying such correlated deficits or a circuit endophenotype would provide a mechanistic point of entry, enhancing both diagnosis and treatment of a given behavioral disorder. We focus on a circuit endophenotype of the thalamic reticular nucleus (TRN) and how its impairment in neurodevelopmental disorders gives rise to a correlated set of readouts across sleep and attention. Because TRN neurons express several disorder-relevant genes identified through genome-wide association studies, exploring the consequences of different TRN disruptions may be of broad translational significance.
Collapse
Affiliation(s)
- Alexandra Krol
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ralf D Wimmer
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michael M Halassa
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
52
|
Yun S, Reynolds RP, Petrof I, White A, Rivera PD, Segev A, Gibson AD, Suarez M, DeSalle MJ, Ito N, Mukherjee S, Richardson DR, Kang CE, Ahrens-Nicklas RC, Soler I, Chetkovich DM, Kourrich S, Coulter DA, Eisch AJ. Stimulation of entorhinal cortex-dentate gyrus circuitry is antidepressive. Nat Med 2018; 24:658-666. [PMID: 29662202 PMCID: PMC5948139 DOI: 10.1038/s41591-018-0002-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 01/26/2018] [Indexed: 12/23/2022]
Abstract
Major Depressive Disorder (MDD) is considered a “circuitopathy”, and brain stimulation therapies hold promise for ameliorating MDD symptoms, including hippocampal dysfunction. It is unknown if stimulation of upstream hippocampal circuitry, such as the entorhinal cortex (Ent), is antidepressive, although Ent stimulation improves learning and memory in lab animals and humans. Here we show molecular targeting (Ent-specific knockdown of a psychosocial stress-induced protein) and chemogenetic stimulation of Ent neurons induce antidepressive-like effects in mice. Mechanistically, we show that Ent stimulation-induced antidepressive-like behavior relies on the generation of new hippocampal neurons. Thus, controlled stimulation of Ent hippocampal afferents is antidepressive via increased hippocampal neurogenesis. These findings emphasize the power and potential of Ent glutamatergic afferent stimulation - previously well known for the ability to influence learning and memory - for MDD treatment.
Collapse
Affiliation(s)
- Sanghee Yun
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Ryan P Reynolds
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Iraklis Petrof
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Alicia White
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Phillip D Rivera
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Pediatrics, Massachusetts General Hospital for Children, Charlestown, MA, USA
| | - Amir Segev
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Adam D Gibson
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Maiko Suarez
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Matthew J DeSalle
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Naoki Ito
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Oriental Medicine Research Center, Kitasato University, Tokyo, Japan
| | - Shibani Mukherjee
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Devon R Richardson
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Catherine E Kang
- Department of Neurology and Clinical Neurological Sciences, Northwestern University, Chicago, IL, USA
| | | | - Ivan Soler
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Dane M Chetkovich
- Department of Neurology and Clinical Neurological Sciences, Northwestern University, Chicago, IL, USA.,Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Saïd Kourrich
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Douglas A Coulter
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Amelia J Eisch
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. .,Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA. .,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
53
|
Liver X receptor β in the hippocampus: A potential novel target for the treatment of major depressive disorder? Neuropharmacology 2018; 135:514-528. [PMID: 29654801 DOI: 10.1016/j.neuropharm.2018.04.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 04/09/2018] [Accepted: 04/10/2018] [Indexed: 12/13/2022]
Abstract
Liver X receptors (LXRs), including LXRα and LXRβ isoforms, have been implicated in multiple physiological functions including promoting neurogenesis, improving synaptic plasticity, preventing neurodegeneration, inhibiting inflammation as well as regulating cholesterol metabolism. However, a potential role of LXRs in the treatment of major depressive disorder (MDD) has never been investigated previously. Our present results demonstrated that levels of hippocampal LXRβ but not LXRα were down-regulated in rats exposed to chronic unpredictable stress (CUS) and were negatively correlated with the severity of CUS-induced depressive-like behaviors. Furthermore, rats with LXRβ knockdown by short hairpin RNA (shRNA) in hippocampus displayed depressive-like behaviors and impaired hippocampal neurogenesis similar to those observed after CUS exposure. Conversely, LXRs activation by GW3965 (GW), a synthetic dual agonist for both LXRα and LXRβ isoforms, could improve depression-like behaviors and reverse the impaired hippocampal neurogenesis in rats exposed to CUS. LXRβ knockdown by shRNA completely abrogated the antidepressant and hippocampal neurogenesis-promoting effects of GW, suggesting that LXRβ isoform mediated the antidepressant and hippocampal neurogenesis-promoting effects of the LXRα/β dual agonist. However, ablation of hippocampal neurogenesis with x-irradiation only partly but not completely abolished the antidepressant effects of GW in the behavioral tests, implying that the antidepressant effects mediated by LXRβ isoform are likely through both neurogenesis-dependent and -independent pathways. Thus, our findings suggest that LXRβ activation may represent a potential novel target for the treatment of MDD and also provide a novel insight into the underlying mechanisms of MDD.
Collapse
|
54
|
Kondo M, Koyama Y, Nakamura Y, Shimada S. A novel 5HT3 receptor-IGF1 mechanism distinct from SSRI-induced antidepressant effects. Mol Psychiatry 2018; 23:833-842. [PMID: 28439104 DOI: 10.1038/mp.2017.87] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 03/16/2017] [Accepted: 03/21/2017] [Indexed: 01/14/2023]
Abstract
Depression is a common mental disorder affecting around 350 million people worldwide. Although selective serotonin reuptake inhibitors (SSRIs) are the most widely used antidepressants, a significant proportion of depressed patients do not achieve remission with SSRIs. In this study, we show that a serotonin type 3 receptor (5HT3R) agonist induces antidepressant effects as well as hippocampal neurogenesis independent of fluoxetine (a commonly used SSRI). Notably, our histological analysis reveals that 5HT3R and insulin-like growth factor 1 (IGF1) are expressed in the same neurons in the subgranular zone of the hippocampal dentate gyrus. Furthermore, our in vivo microdialysis analysis shows that 5HT3R regulates hippocampal extracellular IGF1 levels, and we also show that 5HT3R-dependent hippocampal neurogenesis is mediated by increased IGF1 levels. Altogether, our findings suggest a novel 5HT3R-IGF1 mechanism that is distinct from fluoxetine-induced responses and that provides a new therapeutic target for depression, especially bringing significant benefits for SSRI-resistant depressed patients.
Collapse
Affiliation(s)
- M Kondo
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Y Koyama
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Y Nakamura
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - S Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
55
|
Cameron HA, Schoenfeld TJ. Behavioral and structural adaptations to stress. Front Neuroendocrinol 2018; 49:106-113. [PMID: 29421158 PMCID: PMC5963997 DOI: 10.1016/j.yfrne.2018.02.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/20/2018] [Accepted: 02/03/2018] [Indexed: 10/18/2022]
Abstract
Unpredictable aversive experiences, or stressors, lead to changes in depression- and anxiety-related behavior and to changes in hippocampal structure including decreases in adult neurogenesis, granule cell and pyramidal cell dendritic morphology, and volume. Here we review the relationship between these behavioral and structural changes and discuss the possibility that these changes may be largely adaptive. Specifically, we suggest that new neurons in the dentate gyrus enhance behavioral adaptability to changes in the environment, biasing behavior in novel situations based on previous experience with stress. Conversely, atrophy-like changes in the hippocampus and decreased adult neurogenesis following chronic stress may serve to limit stress responses and stabilize behavior during chronic stress.
Collapse
Affiliation(s)
- Heather A Cameron
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Timothy J Schoenfeld
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
56
|
Sood A, Chaudhari K, Vaidya VA. Acute stress evokes sexually dimorphic, stressor-specific patterns of neural activation across multiple limbic brain regions in adult rats. Stress 2018; 21:136-150. [PMID: 29316846 DOI: 10.1080/10253890.2017.1422488] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Stress enhances the risk for psychiatric disorders such as anxiety and depression. Stress responses vary across sex and may underlie the heightened vulnerability to psychopathology in females. Here, we examined the influence of acute immobilization stress (AIS) and a two-day short-term forced swim stress (FS) on neural activation in multiple cortical and subcortical brain regions, implicated as targets of stress and in the regulation of neuroendocrine stress responses, in male and female rats using Fos as a neural activity marker. AIS evoked a sex-dependent pattern of neural activation within the cingulate and infralimbic subdivisions of the medial prefrontal cortex (mPFC), lateral septum (LS), habenula, and hippocampal subfields. The degree of neural activation in the mPFC, LS, and habenula was higher in males. Female rats exhibited reduced Fos positive cell numbers in the dentate gyrus hippocampal subfield, an effect not observed in males. We addressed whether the sexually dimorphic neural activation pattern noted following AIS was also observed with the short-term stress of FS. In the paraventricular nucleus of the hypothalamus and the amygdala, FS similar to AIS resulted in robust increases in neural activation in both sexes. The pattern of neural activation evoked by FS was distinct across sexes, with a heightened neural activation noted in the prelimbic mPFC subdivision and hippocampal subfields in females and differed from the pattern noted with AIS. This indicates that the sex differences in neural activation patterns observed within stress-responsive brain regions are dependent on the nature of stressor experience.
Collapse
Affiliation(s)
- Ankit Sood
- a Department of Biological Sciences , Tata Institute of Fundamental Research , Mumbai , Maharashtra , India
| | - Karina Chaudhari
- a Department of Biological Sciences , Tata Institute of Fundamental Research , Mumbai , Maharashtra , India
| | - Vidita A Vaidya
- a Department of Biological Sciences , Tata Institute of Fundamental Research , Mumbai , Maharashtra , India
| |
Collapse
|
57
|
Dong J, Zhou Q, Wei Z, Yan S, Sun F, Cai X. Protein kinase A mediates scopolamine-induced mTOR activation and an antidepressant response. J Affect Disord 2018; 227:633-642. [PMID: 29174736 DOI: 10.1016/j.jad.2017.11.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/11/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Clinical reports have shown that scopolamine produces a rapid (3-4 d) and potent anti-depressive response without severe adverse effects. Animal experiments have proven that scopolamine induces mTOR pathway activation in an AMPAR dependent manner. The present study aimed to determine the role of PKA in scopolamine-induced potentiation of AMPAR, as well as in mTOR pathway activation and rapid antidepressant effects. METHODS We utilized electrophysiological recording, Western blotting, and behavior tests to examine the effects of scopolamine, the selective M2 cholinergic receptor antagonist methoctramine, and H89, a PKA specific inhibitor on AMPAR potentiation, mTOR pathway activation, and behavioral responses in a rat depression model of learned helplessness. RESULTS Scopolamine (1μM) rapidly increased AMPAR-fEPSP amplitudes and membrane GluA1 expression in CA1 region of hippocampal slices, both of which were abolished by H89. Moreover, scopolamine promoted AMPAR phosphorylation on GluA1 ser845, a PKA site involved in GluA1 membrane insertion. H89 disrupted both GluA1 ser845 phosphorylation and mTOR activation, as well as the antidepressant effects of scopolamine as determined via forced swim test. Additionally, methoctramine mimicked the effects of scopolamine on phosphorylation and counter-depressive action in a PKA-dependent manner. LIMITATIONS Only one test was used to evaluate depressive behavior, and gene knock-out rats were not yet utilized to refine our hypotheses. CONCLUSION Our findings revealed that PKA pathway is necessary for scopolamine-induced synaptic plasticity and mTOR pathway activation, and indicated that a potential M2-PKA mechanism underlies scopolamine's antidepressant effects. Such findings suggest that GluA1 ser845 phosphorylation may be a trigger event for scopolamine's actions, and that PKA may represent a novel target for the treatment of depressive symptoms.
Collapse
Affiliation(s)
- Jianyang Dong
- Institute of Neurosciences, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qinji Zhou
- Institute of Neurosciences, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhisheng Wei
- Department of Neurology, School of Clinical Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Shi Yan
- Institute of Neurosciences, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fangfang Sun
- Institute of Neurosciences, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiang Cai
- Institute of Neurosciences, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Department of Physiology, Southern Illinois University, Carbondale, IL, USA.
| |
Collapse
|
58
|
Ryu S, Kim HG, Kim JY, Kim SY, Cho KO. Hericium erinaceus Extract Reduces Anxiety and Depressive Behaviors by Promoting Hippocampal Neurogenesis in the Adult Mouse Brain. J Med Food 2017; 21:174-180. [PMID: 29091526 DOI: 10.1089/jmf.2017.4006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Versatile biological activities of Hericium erinaceus (HE) have been reported in many brain diseases. However, roles of HE in major psychiatric disorders such as depression and anxiety remain to be investigated. Therefore, we evaluated whether HE could reduce anxiety and depressive behaviors in the adult mouse and its underlying mechanisms. Male C57BL/6 mice were administered HE (20 or 60 mg/kg, p.o.) or saline once a day for 4 weeks. Open field and tail suspension tests were performed 30 min after the last administration of HE, followed by forced swim test 2 days later. We found that chronic administration of HE showed anxiolytic and antidepressant-like effects. To elucidate possible mechanisms, proliferative activity of the hippocampal progenitor cells was assessed by immunohistochemistry of proliferating cell nuclear antigen (PCNA) and Ki67. Moreover, to evaluate neuronal survival in the dentate gyrus, 5-bromo-2'-deoxyuridine (BrdU) (120 mg/kg, i.p.) was given at the first day of HE administration, followed by isolation of the brains 4 weeks later. HE (60 mg/kg) increased the number of PCNA- and Ki67-positive cells in the subgranular zone of the hippocampus, indicating increased proliferation of hippocampal progenitors. In addition, BrdU- and BrdU/NeuN-positive cells in the dentate gyrus were significantly increased when treated with HE (60 mg/kg) compared with the saline-treated group, demonstrating enhanced neurogenesis by HE treatment. Taken together, the results indicate that chronic HE administration can exert anxiolytic and antidepressant-like effects, possibly by enhancing adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Sun Ryu
- 1 Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea , Seoul, Korea
| | - Hyoun Geun Kim
- 1 Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea , Seoul, Korea
| | - Joo Youn Kim
- 2 Division of New Health Technology Assessment, National Evidence-Based Healthcare Collaborating Agency , Seoul, Korea
| | - Seong Yun Kim
- 1 Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea , Seoul, Korea
| | - Kyung-Ok Cho
- 1 Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea , Seoul, Korea
| |
Collapse
|
59
|
Eliwa H, Belzung C, Surget A. Adult hippocampal neurogenesis: Is it the alpha and omega of antidepressant action? Biochem Pharmacol 2017; 141:86-99. [DOI: 10.1016/j.bcp.2017.08.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 08/04/2017] [Indexed: 01/08/2023]
|
60
|
Neural mechanisms underlying GABAergic regulation of adult hippocampal neurogenesis. Cell Tissue Res 2017; 371:33-46. [PMID: 28948349 DOI: 10.1007/s00441-017-2668-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/01/2017] [Indexed: 12/25/2022]
Abstract
Within the dentate gyrus of the adult hippocampus is the subgranular zone, which contains a neurogenic niche for radial-glia like cells, the most primitive neural stem cells in the adult brain. The quiescence of neural stem cells is maintained by tonic gamma-aminobutyric acid (GABA) released from local interneurons. Once these cells differentiate into neural progenitor cells, GABA continues to regulate their development into mature granule cells, the principal cell type of the dentate gyrus. Here, we review the role of GABA circuits, signaling, and receptors in regulating development of adult-born cells, as well as the molecular players that modulate GABA signaling. Furthermore, we review recent findings linking dysregulation of adult hippocampal neurogenesis to the altered GABAergic circuitry and signaling under various pathological conditions.
Collapse
|
61
|
Neuregulin and Dopamine D4 Receptors Contribute Independently to Depotentiation of Schaffer Collateral LTP by Temperoammonic Path Stimulation. eNeuro 2017; 4:eN-NWR-0176-17. [PMID: 28828402 PMCID: PMC5563842 DOI: 10.1523/eneuro.0176-17.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/14/2017] [Accepted: 08/07/2017] [Indexed: 11/21/2022] Open
Abstract
Prior studies have found that dopamine (DA), acting at D4 receptors, and neuregulin (NRG), likely acting at ErbB4 receptors, are involved in a form of depotentiation of long-term potentiation (LTP) at Schaffer collateral (SC) synapses in the hippocampus. Furthermore, DA and NRG actions are intertwined in that NRG induces DA release. We previously found that low-frequency stimulation (LFS) of temperoammonic (TA) inputs to area CA1 also depotentiates previously established SC LTP through a complex signaling pathway involving endocannabinoids, GABA, adenosine, and mitogen-activated protein kinases (MAPKs), but not glutamate. In the present studies, we found that TA-induced SC depotentiation in hippocampal slices from Sprague-Dawley albino rats also involves activation of both D4 receptors and NRG-activated ErbB receptors, but that the roles of these two modulator systems are independent with D4 receptor antagonism failing to alter chemical depotentiation by NRG1β. Furthermore, a selective D4 receptor agonist was unable to depotentiate SC LTP when administered alone, suggesting that D4 receptor activation is necessary but not sufficient for TA-induced SC depotentiation. Chemical depotentiation by NRG1β was inhibited by a Pan-ErbB antagonist and by picrotoxin (PTX), an antagonist of GABA-A receptors (GABAARs), indicating that NRG likely promotes SC depotentiation via effects on GABA and interneurons. These findings have implications for understanding the role of DA and NRG in cognitive dysfunction associated with neuropsychiatric illnesses.
Collapse
|
62
|
Phillips C. Brain-Derived Neurotrophic Factor, Depression, and Physical Activity: Making the Neuroplastic Connection. Neural Plast 2017; 2017:7260130. [PMID: 28928987 PMCID: PMC5591905 DOI: 10.1155/2017/7260130] [Citation(s) in RCA: 252] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/18/2017] [Indexed: 12/14/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a neurotrophin that is vital to the survival, growth, and maintenance of neurons in key brain circuits involved in emotional and cognitive function. Convergent evidence indicates that neuroplastic mechanisms involving BDNF are deleteriously altered in major depressive disorder (MDD) and animal models of stress. Herein, clinical and preclinical evidence provided that stress-induced depressive pathology contributes to altered BDNF level and function in persons with MDD and, thereby, disruptions in neuroplasticity at the regional and circuit level. Conversely, effective therapeutics that mitigate depressive-related symptoms (e.g., antidepressants and physical activity) optimize BDNF in key brain regions, promote neuronal health and recovery of function in MDD-related circuits, and enhance pharmacotherapeutic response. A greater knowledge of the interrelationship between BDNF, depression, therapeutic mechanisms of action, and neuroplasticity is important as it necessarily precedes the derivation and deployment of more efficacious treatments.
Collapse
|
63
|
Castrén E, Antila H. Neuronal plasticity and neurotrophic factors in drug responses. Mol Psychiatry 2017; 22:1085-1095. [PMID: 28397840 PMCID: PMC5510719 DOI: 10.1038/mp.2017.61] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 02/13/2017] [Accepted: 02/16/2017] [Indexed: 02/07/2023]
Abstract
Neurotrophic factors, particularly brain-derived neurotrophic factor (BDNF) and other members of the neurotrophin family, are central mediators of the activity-dependent plasticity through which environmental experiences, such as sensory information are translated into the structure and function of neuronal networks. Synthesis, release and action of BDNF is regulated by neuronal activity and BDNF in turn leads to trophic effects such as formation, stabilization and potentiation of synapses through its high-affinity TrkB receptors. Several clinically available drugs activate neurotrophin signaling and neuronal plasticity. In particular, antidepressant drugs rapidly activate TrkB signaling and gradually increase BDNF expression, and the behavioral effects of antidepressants are mediated by and dependent on BDNF signaling through TrkB at least in rodents. These findings indicate that antidepressants, widely used drugs, effectively act as TrkB activators. They further imply that neuronal plasticity is a central mechanism in the action of antidepressant drugs. Indeed, it was recently discovered that antidepressants reactivate a state of plasticity in the adult cerebral cortex that closely resembles the enhanced plasticity normally observed during postnatal critical periods. This state of induced plasticity, known as iPlasticity, allows environmental stimuli to beneficially reorganize networks abnormally wired during early life. iPlasticity has been observed in cortical as well as subcortical networks and is induced by several pharmacological and non-pharmacological treatments. iPlasticity is a new pharmacological principle where drug treatment and rehabilitation cooperate; the drug acts permissively to enhance plasticity and rehabilitation provides activity to guide the appropriate wiring of the plastic network. Optimization of iPlastic drug treatment with novel means of rehabilitation may help improve the efficacy of available drug treatments and expand the use of currently existing drugs into new indications.
Collapse
|
64
|
Mendez-David I, Guilloux JP, Papp M, Tritschler L, Mocaer E, Gardier AM, Bretin S, David DJ. S 47445 Produces Antidepressant- and Anxiolytic-Like Effects through Neurogenesis Dependent and Independent Mechanisms. Front Pharmacol 2017; 8:462. [PMID: 28769796 PMCID: PMC5515821 DOI: 10.3389/fphar.2017.00462] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 06/28/2017] [Indexed: 12/20/2022] Open
Abstract
Glutamatergic dysfunctions are observed in the pathophysiology of depression. The glutamatergic synapse as well as the AMPA receptor’s (AMPAR) activation may represent new potential targets for therapeutic intervention in the context of major depressive disorders. S 47445 is a novel AMPARs positive allosteric modulator (AMPA-PAM) possessing procognitive, neurotrophic properties and enhancing synaptic plasticity. Here, we investigated the antidepressant/anxiolytic-like effects of S 47445 in a mouse model of anxiety/depression based on chronic corticosterone administration (CORT) and in the Chronic Mild Stress (CMS) model in rats. Four doses of S 47445 (0.3 to 10 mg/kg, oral route, 4 and 5 weeks, respectively) were assessed in both models. In mouse, behavioral effects were tested in various anxiety-and depression-related behaviors : the elevated plus maze (EPM), open field (OF), splash test (ST), forced swim test (FST), tail suspension test (TST), fur coat state and novelty suppressed feeding (NSF) as well as on hippocampal neurogenesis and dendritic arborization in comparison to chronic fluoxetine treatment (18 mg/kg, p.o.). In rats, behavioral effects of S 47445 were monitored using sucrose consumption and compared to those of imipramine or venlafaxine (10 mg/kg, i.p.) during the whole treatment period and after withdrawal of treatments. In a mouse model of genetic ablation of hippocampal neurogenesis (GFAP-Tk model), neurogenesis dependent/independent effects of chronic S 47445 treatment were tested, as well as BDNF hippocampal expression. S 47445 reversed CORT-induced depressive-like state by increasing grooming duration and reversing coat state’s deterioration. S 47445 also decreased the immobility duration in TST and FST. The highest doses (3 and 10 mg/kg) seem the most effective for antidepressant-like activity in CORT mice. Furthermore, S 47445 significantly reversed the anxiety phenotype observed in OF (at 1 mg/kg) and EPM (from 1 mg/kg). In the CMS rat model, S 47445 (from 1 mg/kg) demonstrated a rapid onset of effect on anhedonia compared to venlafaxine and imipramine. In the CORT model, S 47445 demonstrated significant neurogenic effects on proliferation, survival and maturation of hippocampal newborn neurons at doses inducing an antidepressant-like effect. It also corrected CORT-induced deficits of growth and arborization of dendrites. Finally, the antidepressant/anxiolytic-like activities of S 47445 required adult hippocampal neurogenesis in the novelty suppressed feeding test contrary to OF, EPM and ST. The observed increase in hippocampal BDNF levels could be one of the mechanisms of S 47445 responsible for the adult hippocampal neurogenesis increase. Altogether, S 47445 displays robust antidepressant-anxiolytic-like properties after chronic administration through neurogenesis dependent/independent mechanisms and neuroplastic activities. The AMPA-PAM S 47445 could have promising therapeutic potential for the treatment of major depressive disorders or generalized anxiety disorders.
Collapse
Affiliation(s)
- Indira Mendez-David
- CESP/UMRS-1178, Faculté de Pharmacie, Institut National de la Santé et de la Recherche Médicale, Université Paris-Sud - Université Paris-SaclayChatenay-Malabry, France
| | - Jean-Philippe Guilloux
- CESP/UMRS-1178, Faculté de Pharmacie, Institut National de la Santé et de la Recherche Médicale, Université Paris-Sud - Université Paris-SaclayChatenay-Malabry, France
| | - Mariusz Papp
- Institute of Pharmacology, Polish Academy of SciencesKrakow, Poland
| | - Laurent Tritschler
- CESP/UMRS-1178, Faculté de Pharmacie, Institut National de la Santé et de la Recherche Médicale, Université Paris-Sud - Université Paris-SaclayChatenay-Malabry, France
| | | | - Alain M Gardier
- CESP/UMRS-1178, Faculté de Pharmacie, Institut National de la Santé et de la Recherche Médicale, Université Paris-Sud - Université Paris-SaclayChatenay-Malabry, France
| | - Sylvie Bretin
- Institut de Recherches Internationales ServierSuresnes, France
| | - Denis J David
- CESP/UMRS-1178, Faculté de Pharmacie, Institut National de la Santé et de la Recherche Médicale, Université Paris-Sud - Université Paris-SaclayChatenay-Malabry, France
| |
Collapse
|
65
|
Abstract
Adult hippocampal neurogenesis (AHN) occurs in humans and every other mammalian species examined. Evidence that AHN is stimulated by a variety of treatments and behaviors with anxiolytic properties has sparked interest in harnessing AHN to treat anxiety disorders. However, relatively little is known about the mechanisms through which AHN modulates fear and anxiety. In this review, we consider evidence that AHN modulates fear and anxiety by altering the processing of and memory for traumatic experiences. Based on studies of the role of AHN in Pavlovian fear conditioning, we conclude that AHN modulates the consequences of aversive experience by influencing 1) the efficiency of hippocampus-dependent memory acquisition; 2) generalization of hippocampal fear memories; 3) long-term retention of hippocampal aversive memories; and 4) the nonassociative effects of acute aversive experience. The preclinical literature suggests that stimulation of AHN is likely to have therapeutically relevant consequences, including reduced generalization and long-term retention of aversive memories. However, the literature also identifies four caveats that must be addressed if AHN-based therapies are to achieve therapeutic benefits without significant side effects.
Collapse
Affiliation(s)
- Michael R Drew
- Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, TX, USA.
| | - Kylie A Huckleberry
- Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
66
|
Abstract
Depression is a polygenic and highly complex psychiatric disorder that remains a major burden on society. Antidepressants, such as selective serotonin reuptake inhibitors (SSRIs), are some of the most commonly prescribed drugs worldwide. In this review, we will discuss the evidence that links serotonin and serotonin receptors to the etiology of depression and the mechanisms underlying response to antidepressant treatment. We will then revisit the role of serotonin in three distinct hypotheses that have been proposed over the last several decades to explain the pathophysiology of depression: the monoamine, neurotrophic, and neurogenic hypotheses. Finally, we will discuss how recent studies into serotonin receptors have implicated specific neural circuitry in mediating the antidepressant response, with a focus being placed on the hippocampus.
Collapse
Affiliation(s)
- Christine N Yohn
- Department of Psychology, Behavioral & Systems Neuroscience Area, Rutgers, The State University of New Jersey, 152 Frelinghuysen Rd., Room 215, Piscataway, NJ, 08816, USA
| | - Mark M Gergues
- Department of Psychology, Behavioral & Systems Neuroscience Area, Rutgers, The State University of New Jersey, 152 Frelinghuysen Rd., Room 215, Piscataway, NJ, 08816, USA
| | - Benjamin Adam Samuels
- Department of Psychology, Behavioral & Systems Neuroscience Area, Rutgers, The State University of New Jersey, 152 Frelinghuysen Rd., Room 215, Piscataway, NJ, 08816, USA.
| |
Collapse
|
67
|
Rivnay J, Wang H, Fenno L, Deisseroth K, Malliaras GG. Next-generation probes, particles, and proteins for neural interfacing. SCIENCE ADVANCES 2017; 3:e1601649. [PMID: 28630894 PMCID: PMC5466371 DOI: 10.1126/sciadv.1601649] [Citation(s) in RCA: 224] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 04/18/2017] [Indexed: 05/18/2023]
Abstract
Bidirectional interfacing with the nervous system enables neuroscience research, diagnosis, and therapy. This two-way communication allows us to monitor the state of the brain and its composite networks and cells as well as to influence them to treat disease or repair/restore sensory or motor function. To provide the most stable and effective interface, the tools of the trade must bridge the soft, ion-rich, and evolving nature of neural tissue with the largely rigid, static realm of microelectronics and medical instruments that allow for readout, analysis, and/or control. In this Review, we describe how the understanding of neural signaling and material-tissue interactions has fueled the expansion of the available tool set. New probe architectures and materials, nanoparticles, dyes, and designer genetically encoded proteins push the limits of recording and stimulation lifetime, localization, and specificity, blurring the boundary between living tissue and engineered tools. Understanding these approaches, their modality, and the role of cross-disciplinary development will support new neurotherapies and prostheses and provide neuroscientists and neurologists with unprecedented access to the brain.
Collapse
Affiliation(s)
- Jonathan Rivnay
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Palo Alto Research Center, Palo Alto, CA 94304, USA
- Corresponding author.
| | - Huiliang Wang
- Departments of Bioengineering and Psychiatry, Stanford University, Stanford, CA 94305, USA
| | - Lief Fenno
- Departments of Bioengineering and Psychiatry, Stanford University, Stanford, CA 94305, USA
| | - Karl Deisseroth
- Departments of Bioengineering and Psychiatry, Stanford University, Stanford, CA 94305, USA
| | - George G. Malliaras
- Department of Bioelectronics, École Nationale Supérieure des Mines, CMP-EMSE, MOC, Gardanne 13541, France
| |
Collapse
|
68
|
Campos AC, Fogaça MV, Scarante FF, Joca SRL, Sales AJ, Gomes FV, Sonego AB, Rodrigues NS, Galve-Roperh I, Guimarães FS. Plastic and Neuroprotective Mechanisms Involved in the Therapeutic Effects of Cannabidiol in Psychiatric Disorders. Front Pharmacol 2017; 8:269. [PMID: 28588483 PMCID: PMC5441138 DOI: 10.3389/fphar.2017.00269] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 05/01/2017] [Indexed: 12/25/2022] Open
Abstract
Beneficial effects of cannabidiol (CBD) have been described for a wide range of psychiatric disorders, including anxiety, psychosis, and depression. The mechanisms responsible for these effects, however, are still poorly understood. Similar to clinical antidepressant or atypical antipsychotic drugs, recent findings clearly indicate that CBD, either acutely or repeatedly administered, induces plastic changes. For example, CBD attenuates the decrease in hippocampal neurogenesis and dendrite spines density induced by chronic stress and prevents microglia activation and the decrease in the number of parvalbumin-positive GABA neurons in a pharmacological model of schizophrenia. More recently, it was found that CBD modulates cell fate regulatory pathways such as autophagy and others critical pathways for neuronal survival in neurodegenerative experimental models, suggesting the potential benefit of CBD treatment for psychiatric/cognitive symptoms associated with neurodegeneration. These changes and their possible association with CBD beneficial effects in psychiatric disorders are reviewed here.
Collapse
Affiliation(s)
- Alline C Campos
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Manoela V Fogaça
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Franciele F Scarante
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Sâmia R L Joca
- Department of Physical and Chemical, School of Pharmaceutical Science of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Amanda J Sales
- Department of Physical and Chemical, School of Pharmaceutical Science of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Felipe V Gomes
- Department of Neuroscience, University of PittsburghPittsburgh, PA, United States
| | - Andreza B Sonego
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Naielly S Rodrigues
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Ismael Galve-Roperh
- Department of Biochemistry and Molecular Biology I, School of Biology, Complutense UniversityMadrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Universitario de Investigación en Neuroquímica and Instituto Ramón y Cajal de Investigación SanitariaMadrid, Spain
| | - Francisco S Guimarães
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| |
Collapse
|
69
|
Imoto Y, Segi-Nishida E, Suzuki H, Kobayashi K. Rapid and stable changes in maturation-related phenotypes of the adult hippocampal neurons by electroconvulsive treatment. Mol Brain 2017; 10:8. [PMID: 28253930 PMCID: PMC5335812 DOI: 10.1186/s13041-017-0288-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 02/22/2017] [Indexed: 12/28/2022] Open
Abstract
Electroconvulsive therapy (ECT) is a highly effective and fast-acting treatment for depression. Despite a long history of clinical use, its mechanism of action remains poorly understood. Recently, a novel cellular mechanism of antidepressant action has been proposed: the phenotype of mature brain neurons is transformed to immature-like one by antidepressant drug treatments. We show here that electroconvulsive stimulation (ECS), an animal model of ECT, causes profound changes in maturation-related phenotypes of neurons in the hippocampal dentate gyrus of adult mice. Single ECS immediately reduced expression of mature neuronal markers in almost entire population of dentate granule cells. After ECS treatments, granule cells showed some of physiological properties characteristic of immature granule cells such as higher somatic intrinsic excitability and smaller frequency facilitation at the detate-to-CA3 synapse. The rapid downregulation of maturation markers was suppressed by antagonizing glutamate NMDA receptors, but not by perturbing the serotonergic system. While single ECS caused short-lasting effects, repeated ECS induced stable changes in the maturation-related phenotypes lasting more than 2 weeks along with enhancement of synaptic excitation of granule cells. Augmentation of synaptic inhibition or blockade of NMDA receptors after repeated ECS facilitated regaining the initial mature phenotype, suggesting a role for endogenous neuronal excitation in maintaining the altered maturation-related phenotype probably via NMDA receptor activation. These results suggest that brief neuronal activation by ECS induces "dematuration" of the mature granule cells and that enhanced endogenous excitability is likely to support maintenance of such a demature state. The global increase in neuronal excitability accompanying this process may be relevant to the high efficacy of ECT.
Collapse
Affiliation(s)
- Yuki Imoto
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Eri Segi-Nishida
- Center for Integrative Education in Pharmacy and Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan. .,Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Katsushika-ku, Tokyo, Japan.
| | - Hidenori Suzuki
- Department of Pharmacology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyō, Tokyo, Japan.,Japan Science and Technology Agency, Core Research for Evolutional Science and Technology, Saitama, Japan
| | - Katsunori Kobayashi
- Department of Pharmacology, Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyō, Tokyo, Japan. .,Japan Science and Technology Agency, Core Research for Evolutional Science and Technology, Saitama, Japan.
| |
Collapse
|
70
|
Zhang K, Xu T, Yuan Z, Wei Z, Yamaki VN, Huang M, Huganir RL, Cai X. Essential roles of AMPA receptor GluA1 phosphorylation and presynaptic HCN channels in fast-acting antidepressant responses of ketamine. Sci Signal 2016; 9:ra123. [PMID: 27965425 DOI: 10.1126/scisignal.aai7884] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Although the molecular mechanism is not clear, the clinically tested drug ketamine has rapid antidepressant action that does not require the multiple weeks of treatment needed for other antidepressant drugs to have an effect. We showed that ketamine potentiated Schaffer collateral-CA1 cell excitatory synaptic transmission in hippocampal slice preparations from rodents and enhanced the phosphorylation of the GluA1 subunit on Ser845 of the AMPA-type glutamate receptor in the hippocampal area CA1. These effects persisted when γ-aminobutyric acid (GABA) receptors were pharmacologically blocked. Ketamine reduced behavioral despair in wild-type mice but had no effect in GluA1 S845A knock-in mutant mice. Presynaptic (CA3 pyramidal cell), but not postsynaptic (CA1 pyramidal cell), deletion of N-methyl-d-aspartate (NMDA)-type glutamate receptors eliminated the ketamine-induced enhancement of excitatory synaptic transmission in hippocampal slices and the antidepressant actions of ketamine in mice. The synaptic and behavioral actions of ketamine were completely occluded by inhibition or deletion of the hyperpolarization-activated cyclic nucleotide-gated channel 1 (HCN1). Our results implicate presynaptic NMDA receptor inhibition followed by reduced activity of presynaptic HCN1 channels, which would result in an increase in glutamate release and postsynaptic glutamate receptor activity, as a mechanism of ketamine action. These data provide a mechanism for changes in synaptic activity that could explain the fast-acting antidepressant effects of this drug.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA
| | - Ting Xu
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA.,The Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang Road, Guangzhou, Guangdong 51030, China
| | - Zhongmin Yuan
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA
| | - Zhisheng Wei
- The Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang Road, Guangzhou, Guangdong 51030, China
| | - Vitor Nagai Yamaki
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA
| | - Mingfa Huang
- The Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang Road, Guangzhou, Guangdong 51030, China
| | - Richard L Huganir
- The Solomon H. Snyder Department of Neuroscience, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Xiang Cai
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA.
| |
Collapse
|
71
|
Sweet TB, Hurley SD, Wu MD, Olschowka JA, Williams JP, O'Banion MK. Neurogenic Effects of Low-Dose Whole-Body HZE (Fe) Ion and Gamma Irradiation. Radiat Res 2016; 186:614-623. [PMID: 27905869 DOI: 10.1667/rr14530.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Understanding the dose-toxicity profile of radiation is critical when evaluating potential health risks associated with natural and man-made sources in our environment. The purpose of this study was to evaluate the effects of low-dose whole-body high-energy charged (HZE) iron (Fe) ions and low-energy gamma exposure on proliferation and differentiation of adult-born neurons within the dentate gyrus of the hippocampus, cells deemed to play a critical role in memory regulation. To determine the dose-response characteristics of the brain to whole-body Fe-ion vs. gamma-radiation exposure, C57BL/6J mice were irradiated with 1 GeV/n Fe ions or a static 137Cs source (0.662 MeV) at doses ranging from 0 to 300 cGy. The neurogenesis was analyzed at 48 h and one month postirradiation. These experiments revealed that whole-body exposure to either Fe ions or gamma radiation leads to: 1. An acute decrease in cell division within the dentate gyrus of the hippocampus, detected at doses as low as 30 and 100 cGy for Fe ions and gamma radiation, respectively; and 2. A reduction in newly differentiated neurons (DCX immunoreactivity) at one month postirradiation, with significant decreases detected at doses as low as 100 cGy for both Fe ions and gamma rays. The data presented here contribute to our understanding of brain responses to whole-body Fe ions and gamma rays and may help inform health-risk evaluations related to systemic exposure during a medical or radiologic/nuclear event or as a result of prolonged space travel.
Collapse
Affiliation(s)
- Tara B Sweet
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Sean D Hurley
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Michael D Wu
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - John A Olschowka
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Jacqueline P Williams
- b Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642.,c Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - M Kerry O'Banion
- aDepartment of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642.,d Neurology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| |
Collapse
|
72
|
Yun S, Reynolds RP, Masiulis I, Eisch AJ. Re-evaluating the link between neuropsychiatric disorders and dysregulated adult neurogenesis. Nat Med 2016; 22:1239-1247. [PMID: 27783068 PMCID: PMC5791154 DOI: 10.1038/nm.4218] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 09/30/2016] [Indexed: 12/11/2022]
Abstract
People diagnosed with neuropsychiatric disorders such as depression, anxiety, addiction or schizophrenia often have dysregulated memory, mood, pattern separation and/or reward processing. These symptoms are indicative of a disrupted function of the dentate gyrus (DG) subregion of the brain, and they improve with treatment and remission. The dysfunction of the DG is accompanied by structural maladaptations, including dysregulation of adult-generated neurons. An increasing number of studies using modern inducible approaches to manipulate new neurons show that the behavioral symptoms in animal models of neuropsychiatric disorders can be produced or exacerbated by the inhibition of DG neurogenesis. Thus, here we posit that the connection between neuropsychiatric disorders and dysregulated DG neurogenesis is beyond correlation or epiphenomenon, and that the regulation of adult-generated DG neurogenesis merits continued and focused attention in the ongoing effort to develop novel treatments for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sanghee Yun
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ryan P Reynolds
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Irene Masiulis
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Amelia J Eisch
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neuroscience and Mahoney Institute of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
73
|
Kitahara Y, Nishi A. Antidepressant-induced changes in synaptic morphology in the mouse dentate gyrus. Nihon Yakurigaku Zasshi 2016; 148:180-184. [PMID: 27725565 DOI: 10.1254/fpj.148.180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
74
|
Sánchez-Vidaña DI, Chan NMJ, Chan AH, Hui KK, Lee S, Chan HY, Law YS, Sze MY, Tsui WCS, Fung TK, Lau BWM, Lai CY. Repeated treatment with oxytocin promotes hippocampal cell proliferation, dendritic maturation and affects socio-emotional behavior. Neuroscience 2016; 333:65-77. [DOI: 10.1016/j.neuroscience.2016.07.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/17/2016] [Accepted: 07/02/2016] [Indexed: 11/27/2022]
|
75
|
Zhou WJ, Xu N, Kong L, Sun SC, Xu XF, Jia MZ, Wang Y, Chen ZY. The antidepressant roles of Wnt2 and Wnt3 in stress-induced depression-like behaviors. Transl Psychiatry 2016; 6:e892. [PMID: 27622936 PMCID: PMC5048193 DOI: 10.1038/tp.2016.122] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/23/2016] [Accepted: 05/09/2016] [Indexed: 12/14/2022] Open
Abstract
Wnts-related signaling pathways have been reported to play roles in the pathogenesis of stress-induced depression-like behaviors. However, there is relatively few direct evidence to indicate the effect of Wnt ligands on this process. Here, we investigated the role of Wnts in mediating chronic restraint stress (CRS)-induced depression-like behaviors. We found that CRS induced a significant decrease in the expression of Wnt2 and Wnt3 in the ventral hippocampus (VH) but not in the dorsal hippocampus. Knocking down Wnt2 or Wnt3 in the VH led to impaired Wnt/β-catenin signaling, neurogenesis deficits and depression-like behaviors. In contrast, overexpression of Wnt2 or Wnt3 reversed CRS-induced depression-like behaviors. Moreover, Wnt2 and Wnt3 activated cAMP response element-binding protein (CREB) and there was CREB-dependent positive feedback between Wnt2 and Wnt3. Finally, fluoxetine treatment increased Wnt2 and Wnt3 levels in the VH and knocking down Wnt2 or Wnt3 abolished the antidepressant effect of fluoxetine. Taken together, our study indicates essential roles for Wnt2 and Wnt3 in CRS-induced depression-like behaviors and antidepressant.
Collapse
Affiliation(s)
- W-J Zhou
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, CAS Center for Excellence in Brain Science and Intelligence Technology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - N Xu
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, CAS Center for Excellence in Brain Science and Intelligence Technology, School of Medicine, Shandong University, Jinan, Shandong, China
- Department of Clinical Laboratory, Second Hospital of Shandong University, Jinan, Shandong, China
| | - L Kong
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, CAS Center for Excellence in Brain Science and Intelligence Technology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - S-C Sun
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, CAS Center for Excellence in Brain Science and Intelligence Technology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - X-F Xu
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, CAS Center for Excellence in Brain Science and Intelligence Technology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - M-Z Jia
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, CAS Center for Excellence in Brain Science and Intelligence Technology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Y Wang
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, CAS Center for Excellence in Brain Science and Intelligence Technology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Z-Y Chen
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, CAS Center for Excellence in Brain Science and Intelligence Technology, School of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
76
|
Baydin S, Yagmurlu K, Tanriover N, Gungor A, Rhoton AL. Microsurgical and Fiber Tract Anatomy of the Nucleus Accumbens. Oper Neurosurg (Hagerstown) 2016; 12:269-288. [DOI: 10.1227/neu.0000000000001133] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 10/04/2015] [Indexed: 11/19/2022] Open
|
77
|
Kang E, Wen Z, Song H, Christian KM, Ming GL. Adult Neurogenesis and Psychiatric Disorders. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a019026. [PMID: 26801682 DOI: 10.1101/cshperspect.a019026] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Psychiatric disorders continue to be among the most challenging disorders to diagnose and treat because there is no single genetic or anatomical locus that is causative for the disease. Current treatments are often blunt tools used to ameliorate the most severe symptoms, at the risk of disrupting functional neural systems. There is a critical need to develop new therapeutic strategies that can target circumscribed functional or anatomical domains of pathology. Adult hippocampal neurogenesis may be one such domain. Here, we review the evidence suggesting that adult hippocampal neurogenesis plays a role in emotional regulation and forms of learning and memory that include temporal and spatial memory encoding and context discrimination, and that its dysregulation is associated with psychiatric disorders, such as affective disorders, schizophrenia, and drug addiction. Further, adult neurogenesis has proven to be an effective model to investigate basic processes of neuronal development and converging evidence suggests that aberrant neural development may be an etiological factor, even in late-onset diseases. Constitutive neurogenesis in the hippocampus of the mature brain reflects large-scale plasticity unique to this region and could be a potential hub for modulation of a subset of cognitive and affective behaviors that are affected by multiple psychiatric disorders.
Collapse
Affiliation(s)
- Eunchai Kang
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Zhexing Wen
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Kimberly M Christian
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Guo-Li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
78
|
Kondo M. Molecular mechanisms of experience-dependent structural and functional plasticity in the brain. Anat Sci Int 2016; 92:1-17. [DOI: 10.1007/s12565-016-0358-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/16/2016] [Indexed: 11/24/2022]
|
79
|
Zhang J, Cai CY, Wu HY, Zhu LJ, Luo CX, Zhu DY. CREB-mediated synaptogenesis and neurogenesis is crucial for the role of 5-HT1a receptors in modulating anxiety behaviors. Sci Rep 2016; 6:29551. [PMID: 27404655 PMCID: PMC4941576 DOI: 10.1038/srep29551] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/17/2016] [Indexed: 02/03/2023] Open
Abstract
Serotonin 1a-receptor (5-HT1aR) has been specifically implicated in the pathogenesis of anxiety. However, the mechanism underlying the role of 5-HT1aR in anxiety remains poorly understood. Here we show in mice that the transcription factor cAMP response element binding protein (CREB) in the hippocampus functions as an effector of 5-HT1aR in modulating anxiety-related behaviors. We generated recombinant lentivirus LV-CREB133-GFP expressing a dominant negative CREB which could not be phosphorylated at Ser133 to specifically reduce CREB activity, and LV-VP16-CREB-GFP expressing a constitutively active fusion protein VP16-CREB which could be phosphorylated by itself to specifically enhance CREB activity. LV-CREB133-GFP neutralized 5-HT1aR agonist-induced up-regulation of synapse density, spine density, dendrite complexity, neurogenesis, and the expression of synapsin and spinophilin, two well-characterized synaptic proteins, and abolished the anxiolytic effect of 5-HT1aR agonist; whereas LV-VP16-CREB-GFP rescued the 5-HT1aR antagonist-induced down-regulation of synapse density, spine density, dendrite complexity, neurogenesis and synapsin and spinophilin expression, and reversed the anxiogenic effect of 5-HT1aR antagonist. The deletion of neurogenesis by irradiation or the diminution of synaptogenesis by knockdown of synapsin expression abolished the anxiolytic effects of both CREB and 5-HT1aR activation. These findings suggest that CREB-mediated hippoacampus structural plasticity is crucial for the role of 5-HT1aR in modulating anxiety-related behaviors.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 210029, China.,Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing 210029, China
| | - Cheng-Yun Cai
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 210029, China
| | - Hai-Yin Wu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 210029, China.,Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing 210029, China
| | - Li-Juan Zhu
- Institute of Neuroscience, Soochow University, Su zhou, China
| | - Chun-Xia Luo
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 210029, China.,Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing 210029, China
| | - Dong-Ya Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 210029, China.,Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing 210029, China.,The key laboratory of human functional genomics of Jiangsu Province, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
80
|
Tannenholz L, Hen R, Kheirbek MA. GluN2B-Containg NMDA Receptors on Adult-Born Granule Cells Contribute to the Antidepressant Action of Fluoxetine. Front Neurosci 2016; 10:242. [PMID: 27303260 PMCID: PMC4885883 DOI: 10.3389/fnins.2016.00242] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/17/2016] [Indexed: 01/09/2023] Open
Abstract
Ablation of adult neurogenesis in mice has revealed that young adult-born granule cells (abGCs) are required for some of the behavioral responses to antidepressants (ADs), yet the mechanism by which abGCs contribute to AD action remains unknown. During their maturation process, these immature neurons exhibit unique properties that could underlie their ability to influence behavioral output. In particular, abGCs in the DG exhibit a period of heightened plasticity 4–6 weeks after birth that is mediated by GluN2B-expressing NMDA receptors. The functional contribution of this critical window to AD responsiveness is unclear. Here, we determined the behavioral and neurogenic responses to the AD fluoxetine (FLX) in mice lacking GluN2B-containing NMDA receptors in abGCs. We found that these mice exhibited an attenuated response to FLX in a neurogenesis-dependent behavioral assay of FLX action, while neurogenesis-independent behaviors were unaffected by GluN2B deletion. In addition, deletion of GluN2B attenuated FLX-induced increases in dendritic complexity of abGCs suggesting that the blunted behavioral efficacy of FLX may be caused by impaired differentiation of young abGCs.
Collapse
Affiliation(s)
- Lindsay Tannenholz
- Department of Pharmacology, Columbia UniversityNew York, NY, USA; Division of Integrative Neuroscience, New York State Psychiatric InstituteNew York, NY, USA
| | - René Hen
- Department of Pharmacology, Columbia UniversityNew York, NY, USA; Division of Integrative Neuroscience, New York State Psychiatric InstituteNew York, NY, USA; Department of Psychiatry, Columbia UniversityNew York, NY, USA; Department of Neuroscience, Columbia UniversityNew York, NY, USA
| | - Mazen A Kheirbek
- Division of Integrative Neuroscience, New York State Psychiatric InstituteNew York, NY, USA; Department of Psychiatry, Columbia UniversityNew York, NY, USA; Department of Psychiatry, University of CaliforniaSan Francisco, CA, USA
| |
Collapse
|
81
|
Effect of zinc supplementation on neuronal precursor proliferation in the rat hippocampus after traumatic brain injury. Exp Neurol 2016; 279:96-103. [DOI: 10.1016/j.expneurol.2016.02.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/16/2016] [Accepted: 02/18/2016] [Indexed: 01/27/2023]
|
82
|
Yun S, Donovan MH, Ross MN, Richardson DR, Reister R, Farnbauch LA, Fischer SJ, Riethmacher D, Gershenfeld HK, Lagace DC, Eisch AJ. Stress-Induced Anxiety- and Depressive-Like Phenotype Associated with Transient Reduction in Neurogenesis in Adult Nestin-CreERT2/Diphtheria Toxin Fragment A Transgenic Mice. PLoS One 2016; 11:e0147256. [PMID: 26795203 PMCID: PMC4721672 DOI: 10.1371/journal.pone.0147256] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 01/02/2016] [Indexed: 01/01/2023] Open
Abstract
Depression and anxiety involve hippocampal dysfunction, but the specific relationship between these mood disorders and adult hippocampal dentate gyrus neurogenesis remains unclear. In both humans with MDD and rodent models of depression, administration of antidepressants increases DG progenitor and granule cell number, yet rodents with induced ablation of DG neurogenesis typically do not demonstrate depressive- or anxiety-like behaviors. The conflicting data may be explained by the varied duration and degree to which adult neurogenesis is reduced in different rodent neurogenesis ablation models. In order to test this hypothesis we examined how a transient–rather than permanent–inducible reduction in neurogenesis would alter depressive- and anxiety-like behaviors. Transgenic Nestin-CreERT2/floxed diphtheria toxin fragment A (DTA) mice (Cre+DTA+) and littermates (Cre+DTA-; control) were given tamoxifen (TAM) to induce recombination and decrease nestin-expressing stem cells and their progeny. The decreased neurogenesis was transient: 12 days post-TAM Cre+DTA+ mice had fewer DG proliferating Ki67+ cells and fewer DCX+ neuroblasts/immature neurons relative to control, but 30 days post-TAM Cre+DTA+ mice had the same DCX+ cell number as control. This ability of DG neurogenesis to recover after partial ablation also correlated with changes in behavior. Relative to control, Cre+DTA+ mice tested between 12–30 days post-TAM displayed indices of a stress-induced anxiety phenotype–longer latency to consume highly palatable food in the unfamiliar cage in the novelty-induced hypophagia test, and a depression phenotype–longer time of immobility in the tail suspension test, but Cre+DTA+ mice tested after 30 days post-TAM did not. These findings suggest a functional association between adult neurogenesis and stress induced anxiety- and depressive-like behaviors, where induced reduction in DCX+ cells at the time of behavioral testing is coupled with stress-induced anxiety and a depressive phenotype, and recovery of DCX+ cell number corresponds to normalization of these behaviors.
Collapse
Affiliation(s)
- Sanghee Yun
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Michael H. Donovan
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Michele N. Ross
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Devon R. Richardson
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Robin Reister
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Laure A. Farnbauch
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Stephanie J. Fischer
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Dieter Riethmacher
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana, Kazakhstan
- Human Development and Health, School of Medicine, Southampton General Hospital, Southampton University, Southampton, United Kingdom
| | - Howard K. Gershenfeld
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Diane C. Lagace
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
- * E-mail: (AJE); (DCL)
| | - Amelia J. Eisch
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
- * E-mail: (AJE); (DCL)
| |
Collapse
|
83
|
Kitahara Y, Ohta K, Hasuo H, Shuto T, Kuroiwa M, Sotogaku N, Togo A, Nakamura KI, Nishi A. Chronic Fluoxetine Induces the Enlargement of Perforant Path-Granule Cell Synapses in the Mouse Dentate Gyrus. PLoS One 2016; 11:e0147307. [PMID: 26788851 PMCID: PMC4720354 DOI: 10.1371/journal.pone.0147307] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/01/2016] [Indexed: 12/27/2022] Open
Abstract
A selective serotonin reuptake inhibitor is the most commonly prescribed antidepressant for the treatment of major depression. However, the mechanisms underlying the actions of selective serotonin reuptake inhibitors are not fully understood. In the dentate gyrus, chronic fluoxetine treatment induces increased excitability of mature granule cells (GCs) as well as neurogenesis. The major input to the dentate gyrus is the perforant path axons (boutons) from the entorhinal cortex (layer II). Through voltage-sensitive dye imaging, we found that the excitatory neurotransmission of the perforant path synapse onto the GCs in the middle molecular layer of the mouse dentate gyrus (perforant path-GC synapse) is enhanced after chronic fluoxetine treatment (15 mg/kg/day, 14 days). Therefore, we further examined whether chronic fluoxetine treatment affects the morphology of the perforant path-GC synapse, using FIB/SEM (focused ion beam/scanning electron microscopy). A three-dimensional reconstruction of dendritic spines revealed the appearance of extremely large-sized spines after chronic fluoxetine treatment. The large-sized spines had a postsynaptic density with a large volume. However, chronic fluoxetine treatment did not affect spine density. The presynaptic boutons that were in contact with the large-sized spines were large in volume, and the volumes of the mitochondria and synaptic vesicles inside the boutons were correlated with the size of the boutons. Thus, the large-sized perforant path-GC synapse induced by chronic fluoxetine treatment contains synaptic components that correlate with the synapse size and that may be involved in enhanced glutamatergic neurotransmission.
Collapse
Affiliation(s)
- Yosuke Kitahara
- Department of Pharmacology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830–0011, Japan
| | - Keisuke Ohta
- Department of Anatomy, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830–0011, Japan
| | - Hiroshi Hasuo
- Department of Physiology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830–0011, Japan
| | - Takahide Shuto
- Department of Pharmacology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830–0011, Japan
| | - Mahomi Kuroiwa
- Department of Pharmacology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830–0011, Japan
| | - Naoki Sotogaku
- Department of Pharmacology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830–0011, Japan
| | - Akinobu Togo
- Department of Anatomy, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830–0011, Japan
| | - Kei-ichiro Nakamura
- Department of Anatomy, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830–0011, Japan
| | - Akinori Nishi
- Department of Pharmacology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830–0011, Japan
- * E-mail:
| |
Collapse
|
84
|
Stepan J, Hladky F, Uribe A, Holsboer F, Schmidt MV, Eder M. High-Speed imaging reveals opposing effects of chronic stress and antidepressants on neuronal activity propagation through the hippocampal trisynaptic circuit. Front Neural Circuits 2015; 9:70. [PMID: 26594153 PMCID: PMC4635222 DOI: 10.3389/fncir.2015.00070] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 10/22/2015] [Indexed: 12/11/2022] Open
Abstract
Antidepressants (ADs) are used as first-line treatment for most stress-related psychiatric disorders. The alterations in brain circuit dynamics that can arise from stress exposure and underlie therapeutic actions of ADs remain, however, poorly understood. Here, enabled by a recently developed voltage-sensitive dye imaging (VSDI) assay in mouse brain slices, we examined the impact of chronic stress and concentration-dependent effects of eight clinically used ADs (belonging to different chemical/functional classes) on evoked neuronal activity propagations through the hippocampal trisynaptic circuitry (HTC: perforant path → dentate gyrus (DG) → area CA3 → area CA1). Exposure of mice to chronic social defeat stress led to markedly weakened activity propagations (“HTC-Waves”). In contrast, at concentrations in the low micromolar range, all ADs, which were bath applied to slices, caused an amplification of HTC-Waves in CA regions (invariably in area CA1). The fast-acting “antidepressant” ketamine, the mood stabilizer lithium, and brain-derived neurotrophic factor (BDNF) exerted comparable enhancing effects, whereas the antipsychotic haloperidol and the anxiolytic diazepam attenuated HTC-Waves. Collectively, we provide direct experimental evidence that chronic stress can depress neuronal signal flow through the HTC and demonstrate shared opposing effects of ADs. Thus, our study points to a circuit-level mechanism of ADs to counteract stress-induced impairment of hippocampal network function. However, the observed effects of ADs are impossible to depend on enhanced neurogenesis.
Collapse
Affiliation(s)
- Jens Stepan
- Max Planck Institute of Psychiatry Munich, Germany ; Department Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry Munich, Germany ; Scientific Core Unit "Electrophysiology and Neuronal Network Dynamics", Max Planck Institute of Psychiatry Munich, Germany ; Clinical Department, Max Planck Institute of Psychiatry Munich, Germany
| | - Florian Hladky
- Max Planck Institute of Psychiatry Munich, Germany ; Department Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry Munich, Germany ; Scientific Core Unit "Electrophysiology and Neuronal Network Dynamics", Max Planck Institute of Psychiatry Munich, Germany
| | - Andrés Uribe
- Max Planck Institute of Psychiatry Munich, Germany ; Department Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry Munich, Germany ; Research Group "Stress Resilience", Max Planck Institute of Psychiatry Munich, Germany
| | - Florian Holsboer
- Max Planck Institute of Psychiatry Munich, Germany ; HMNC GmbH Munich, Germany
| | - Mathias V Schmidt
- Max Planck Institute of Psychiatry Munich, Germany ; Department Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry Munich, Germany ; Research Group "Stress Resilience", Max Planck Institute of Psychiatry Munich, Germany
| | - Matthias Eder
- Max Planck Institute of Psychiatry Munich, Germany ; Department Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry Munich, Germany ; Scientific Core Unit "Electrophysiology and Neuronal Network Dynamics", Max Planck Institute of Psychiatry Munich, Germany
| |
Collapse
|
85
|
Adult Hippocampal Neurogenesis Modulates Fear Learning through Associative and Nonassociative Mechanisms. J Neurosci 2015; 35:11330-45. [PMID: 26269640 DOI: 10.1523/jneurosci.0483-15.2015] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Adult hippocampal neurogenesis is believed to support hippocampus-dependent learning and emotional regulation. These putative functions of adult neurogenesis have typically been studied in isolation, and little is known about how they interact to produce adaptive behavior. We used trace fear conditioning as a model system to elucidate mechanisms through which adult hippocampal neurogenesis modulates processing of aversive experience. To achieve a specific ablation of neurogenesis, we generated transgenic mice that express herpes simplex virus thymidine kinase specifically in neural progenitors and immature neurons. Intracerebroventricular injection of the prodrug ganciclovir caused a robust suppression of neurogenesis without suppressing gliogenesis. Neurogenesis ablation via this method or targeted x-irradiation caused an increase in context conditioning in trace but not delay fear conditioning. Data suggest that this phenotype represents opposing effects of neurogenesis ablation on associative and nonassociative components of fear learning. Arrest of neurogenesis sensitizes mice to nonassociative effects of fear conditioning, as evidenced by increased anxiety-like behavior in the open field after (but not in the absence of) fear conditioning. In addition, arrest of neurogenesis impairs associative trace conditioning, but this impairment can be masked by nonassociative fear. The results suggest that adult neurogenesis modulates emotional learning via two distinct but opposing mechanisms: it supports associative trace conditioning while also buffering against the generalized fear and anxiety caused by fear conditioning. SIGNIFICANCE STATEMENT The role of adult hippocampal neurogenesis in fear learning is controversial, with some studies suggesting neurogenesis is needed for aspects of fear learning and others suggesting it is dispensable. We generated transgenic mice in which neural progenitors can be selectively and inducibly ablated. Our data suggest that adult neurogenesis supports fear learning through two distinct mechanisms: it supports the ability to learn associations between traumatic events (unconditioned stimuli) and predictors (conditioned stimuli) while also buffering against nonassociative, anxiogenic effects of a traumatic experience. As a result, arrest of neurogenesis can enhance or impair learned fear depending on intensity of the traumatic experience and the extent to which it recruits associative versus nonassociative learning.
Collapse
|
86
|
Neuroplasticity-dependent and -independent mechanisms of chronic deep brain stimulation in stressed rats. Transl Psychiatry 2015; 5:e674. [PMID: 26529427 PMCID: PMC5068759 DOI: 10.1038/tp.2015.166] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 09/16/2015] [Accepted: 09/19/2015] [Indexed: 12/24/2022] Open
Abstract
Chronic ventromedial prefrontal cortex (vmPFC) deep brain stimulation (DBS) improves depressive-like behaviour in rats via serotonergic and neurotrophic-related mechanisms. We hypothesise that, in addition to these substrates, DBS-induced increases in hippocampal neurogenesis may also be involved. Our results show that stress-induced behavioural deficits in the sucrose preference test, forced swim test, novelty-suppressed feeding test (NSFT) and elevated plus maze were countered by chronic vmPFC DBS. In addition, stressed rats receiving stimulation had significant increases in hippocampal neurogenesis, PFC and hippocampal brain-derived neurotrophic factor levels. To block neurogenesis, stressed animals given DBS were injected with temozolomide. Such treatment reversed the anxiolytic-like effect of stimulation in the NSFT without significantly affecting performance in other behavioural tests. Taken together, our findings suggest that neuroplastic changes, including neurogenesis, may be involved in specific anxiolytic effects of DBS without affecting its general antidepressant-like response.
Collapse
|
87
|
Malykhin N, Coupland N. Hippocampal neuroplasticity in major depressive disorder. Neuroscience 2015; 309:200-13. [DOI: 10.1016/j.neuroscience.2015.04.047] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 04/17/2015] [Accepted: 04/21/2015] [Indexed: 01/31/2023]
|
88
|
Kondo M, Nakamura Y, Ishida Y, Shimada S. The 5-HT3 receptor is essential for exercise-induced hippocampal neurogenesis and antidepressant effects. Mol Psychiatry 2015; 20:1428-37. [PMID: 25403840 DOI: 10.1038/mp.2014.153] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/03/2014] [Accepted: 10/08/2014] [Indexed: 12/22/2022]
Abstract
Exercise has a variety of beneficial effects on brain structure and function, such as hippocampal neurogenesis, mood and memory. Previous studies have shown that exercise enhances hippocampal neurogenesis, induces antidepressant effects and improves learning behavior. Brain serotonin (5-hydroxytryptamine, 5-HT) levels increase following exercise, and the 5-HT system has been suggested to have an important role in these exercise-induced neuronal effects. However, the precise mechanism remains unclear. In this study, analysis of the 5-HT type 3A receptor subunit-deficient (htr3a(-/-)) mice revealed that lack of the 5-HT type 3 (5-HT3) receptor resulted in loss of exercise-induced hippocampal neurogenesis and antidepressant effects, but not of learning enhancement. Furthermore, stimulation of the 5-HT3 receptor promoted neurogenesis. These findings demonstrate that the 5-HT3 receptor is the critical target of 5-HT action in the brain following exercise, and is indispensable for hippocampal neurogenesis and antidepressant effects induced by exercise. This is the first report of a pivotal 5-HT receptor subtype that has a fundamental role in exercise-induced morphological changes and psychological effects.
Collapse
Affiliation(s)
- M Kondo
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Y Nakamura
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Y Ishida
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - S Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
89
|
Choudhury A, Singh S, Palit G, Shukla S, Ganguly S. Administration of N-acetylserotonin and melatonin alleviate chronic ketamine-induced behavioural phenotype accompanying BDNF-independent and dependent converging cytoprotective mechanisms in the hippocampus. Behav Brain Res 2015; 297:204-12. [PMID: 26475510 DOI: 10.1016/j.bbr.2015.10.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 09/15/2015] [Accepted: 10/08/2015] [Indexed: 12/14/2022]
Abstract
Though growing evidence implicates both melatonin (MLT) and its immediate precursor N-acetylserotonin (NAS) in the regulation of hippocampal neurogenesis, their comparative mechanistic relationship with core behavioural correlates of psychiatric disorders is largely unknown. To address this issue, we investigated the ability of these indoleamines to mitigate the behavioral phenotypes associated with NMDA-receptor (NMDAR) hypofunction in mice. We demonstrated that exogenous MLT and NAS treatments attenuated the NMDAR antagonist (ketamine) induced immobility in the forced swim test (FST) but not the classical striatum-related hyperlocomotor activity phenotype. The MLT/NAS-mediated protection of the phenotype in FST could be correlated to the ability of these indoleamines to counteract the deleterious effects of chronic ketamine on pro-survival molecular events by restoring the activities in MEK-ERK and PI3K-AKT pathways in the hippocampus. MLT seems to modulate these pathways by promoting accumulation of the mature form of BDNF above the control (vehicle-treated) levels, perhaps via MLT receptor-dependent mechanisms and in the process overcoming the ketamine-induced down-regulation of BDNF. In contrast, NAS appears to partly restore the ketamine-induced decrease of BDNF to the control levels. In spite of this fundamental difference in modulating BDNF levels in the upstream events, both MLT and NAS seem to overlap in the TrkB-induced downstream pro-survival mechanisms in the hippocampus, providing protection against NMDAR-hypofunction related cellular events. Perhaps, this also signifies the physiological importance of robust MLT synthesizing machinery that converts serotonin to MLT, in ensuring positive impact on hippocampus-related symptoms in psychiatric disorders.
Collapse
Affiliation(s)
- Arnab Choudhury
- Chronic Disease Biology Group, Institute of Molecular Medicine, 254 Okhla Industrial Estate, Phase-3, New Delhi 110020, India
| | - Seema Singh
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031 U.P, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001, India
| | - Gautam Palit
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031 U.P, India
| | - Shubha Shukla
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031 U.P, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001, India.
| | - Surajit Ganguly
- Chronic Disease Biology Group, Institute of Molecular Medicine, 254 Okhla Industrial Estate, Phase-3, New Delhi 110020, India.
| |
Collapse
|
90
|
Grimonprez A, Raedt R, Portelli J, Dauwe I, Larsen LE, Bouckaert C, Delbeke J, Carrette E, Meurs A, De Herdt V, Boon P, Vonck K. The antidepressant-like effect of vagus nerve stimulation is mediated through the locus coeruleus. J Psychiatr Res 2015; 68:1-7. [PMID: 26228393 DOI: 10.1016/j.jpsychires.2015.05.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 04/02/2015] [Accepted: 05/06/2015] [Indexed: 02/06/2023]
Abstract
It has been shown that vagus nerve stimulation (VNS) has an antidepressant-like effect in the forced swim test. The mechanism of action underlying this effect is incompletely understood, but there is evidence suggesting that the locus coeruleus (LC) may play an important role. In this study, noradrenergic LC neurons were selectively lesioned to test their involvement in the antidepressant-like effect of VNS in the forced swim test. Forced swim test behavior was assessed in rats that were subjected to VNS or sham treatment. In half of the VNS-treated animals, the noradrenergic neurons from the LC were lesioned using the selective neurotoxin DSP-4 [N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride], yielding three experimental arms: sham, VNS and DSP-4-VNS (n = 8 per group). Furthermore, the open field test was performed to evaluate locomotor activity. A dopamine-β-hydroxylase immunostaining was performed to confirm lesioning of noradrenergic LC neurons. VNS significantly reduced the percentage of immobility time in the forced swim test compared to sham treatment (median: 56%, interquartile range: 41% vs. median: 75%, interquartile range: 12%). This antidepressant-like effect of VNS could not be demonstrated in the DSP-4-VNS group (median: 79%, interquartile range: 33%). Locomotor activity in the open field test was not different between the three treatment arms. The absence of hippocampal dopamine-β-hydroxylase immunostaining in the DSP-4-treated rats confirmed the lesioning of noradrenergic neurons originating from the brainstem LC. The results of this study demonstrate that the noradrenergic neurons from the LC play an important role in the antidepressant-like effect of VNS.
Collapse
Affiliation(s)
- Annelies Grimonprez
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology (LCEN3), Ghent University, Department of Neurology, Institute for Neuroscience, Ghent, Belgium.
| | - Robrecht Raedt
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology (LCEN3), Ghent University, Department of Neurology, Institute for Neuroscience, Ghent, Belgium.
| | - Jeanelle Portelli
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology (LCEN3), Ghent University, Department of Neurology, Institute for Neuroscience, Ghent, Belgium; Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Ine Dauwe
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology (LCEN3), Ghent University, Department of Neurology, Institute for Neuroscience, Ghent, Belgium.
| | - Lars Emil Larsen
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology (LCEN3), Ghent University, Department of Neurology, Institute for Neuroscience, Ghent, Belgium.
| | - Charlotte Bouckaert
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology (LCEN3), Ghent University, Department of Neurology, Institute for Neuroscience, Ghent, Belgium.
| | - Jean Delbeke
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology (LCEN3), Ghent University, Department of Neurology, Institute for Neuroscience, Ghent, Belgium.
| | - Evelien Carrette
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology (LCEN3), Ghent University, Department of Neurology, Institute for Neuroscience, Ghent, Belgium.
| | - Alfred Meurs
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology (LCEN3), Ghent University, Department of Neurology, Institute for Neuroscience, Ghent, Belgium.
| | - Veerle De Herdt
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology (LCEN3), Ghent University, Department of Neurology, Institute for Neuroscience, Ghent, Belgium.
| | - Paul Boon
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology (LCEN3), Ghent University, Department of Neurology, Institute for Neuroscience, Ghent, Belgium.
| | - Kristl Vonck
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology (LCEN3), Ghent University, Department of Neurology, Institute for Neuroscience, Ghent, Belgium.
| |
Collapse
|
91
|
Calcitonin gene-related peptide pre-administration acts as a novel antidepressant in stressed mice. Sci Rep 2015; 5:12559. [PMID: 26251188 PMCID: PMC4528222 DOI: 10.1038/srep12559] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 07/03/2015] [Indexed: 12/21/2022] Open
Abstract
Calcitonin gene-related peptide (CGRP) is a neuropeptide that has potent vasodilator properties and is involved in various behavioral disorders. The relationship between CGRP and depression-like behavior is unclear. In this study, we used chronically stressed mice to investigate whether CGRP is involved in depression-like behavior. Each mouse was exposed to restraint and water immersion stress for 15 days. After stress exposure, mice were assessed using behavioral tests: open field test, forced swim test and sucrose preference test. Serum corticosterone levels, hippocampal proliferation and mRNA expression of neurotrophins were measured. After stress exposure, mice exhibited depression-like behavior and decreased CGRP mRNA levels in the hippocampus. Although intracerebroventricular CGRP administration (0.5 nmol) did not alter depression-like behavior after 15-day stress exposure, a single CGRP administration into the brain, before the beginning of the 15-day stress exposure, normalized the behavioral dysfunctions and increased nerve growth factor (Ngf) mRNA levels in stressed mice. Furthermore, in the mouse E14 hippocampal cell line, CGRP treatment induced increased expression of Ngf mRNA. The NGF receptor inhibitor K252a inhibited CGRP's antidepressant-like effects in stressed mice. These results suggest that CGRP expression in the mouse hippocampus is associated with depression-like behavior and changes in Ngf mRNA levels.
Collapse
|
92
|
Wang Y, Ma Y, Hu J, Cheng W, Jiang H, Zhang X, Li M, Ren J, Li X. Prenatal chronic mild stress induces depression-like behavior and sex-specific changes in regional glutamate receptor expression patterns in adult rats. Neuroscience 2015; 301:363-74. [DOI: 10.1016/j.neuroscience.2015.06.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 06/03/2015] [Accepted: 06/04/2015] [Indexed: 12/31/2022]
|
93
|
Erburu M, Cajaleon L, Guruceaga E, Venzala E, Muñoz-Cobo I, Beltrán E, Puerta E, Tordera R. Chronic mild stress and imipramine treatment elicit opposite changes in behavior and in gene expression in the mouse prefrontal cortex. Pharmacol Biochem Behav 2015; 135:227-36. [DOI: 10.1016/j.pbb.2015.06.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 05/28/2015] [Accepted: 06/01/2015] [Indexed: 01/22/2023]
|
94
|
Ramirez S, Liu X, MacDonald CJ, Moffa A, Zhou J, Redondo RL, Tonegawa S. Activating positive memory engrams suppresses depression-like behaviour. Nature 2015; 522:335-9. [PMID: 26085274 PMCID: PMC5583720 DOI: 10.1038/nature14514] [Citation(s) in RCA: 220] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 05/01/2015] [Indexed: 12/17/2022]
Abstract
Stress is considered a potent environmental risk factor for many behavioural abnormalities, including anxiety and mood disorders. Animal models can exhibit limited but quantifiable behavioural impairments resulting from chronic stress, including deficits in motivation, abnormal responses to behavioural challenges, and anhedonia. The hippocampus is thought to negatively regulate the stress response and to mediate various cognitive and mnemonic aspects of stress-induced impairments, although the neuronal underpinnings sufficient to support behavioural improvements are largely unknown. Here we acutely rescue stress-induced depression-related behaviours in mice by optogenetically reactivating dentate gyrus cells that were previously active during a positive experience. A brain-wide histological investigation, coupled with pharmacological and projection-specific optogenetic blockade experiments, identified glutamatergic activity in the hippocampus-amygdala-nucleus-accumbens pathway as a candidate circuit supporting the acute rescue. Finally, chronically reactivating hippocampal cells associated with a positive memory resulted in the rescue of stress-induced behavioural impairments and neurogenesis at time points beyond the light stimulation. Together, our data suggest that activating positive memories artificially is sufficient to suppress depression-like behaviours and point to dentate gyrus engram cells as potential therapeutic nodes for intervening with maladaptive behavioural states.
Collapse
Affiliation(s)
- Steve Ramirez
- RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | - Christopher J MacDonald
- RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Anthony Moffa
- RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Joanne Zhou
- RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Roger L Redondo
- 1] RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA [2] Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Susumu Tonegawa
- 1] RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA [2] Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
95
|
Abstract
The wide spectrum of disruptions that characterizes major depressive disorder (MDD) and bipolar disorder (BD) highlights the difficulties researchers are posed with as they try to mimic these disorders in the laboratory. Nonetheless, numerous attempts have been made to create rodent models of mood disorders or at least models of the symptoms of MDD and BD. Present antidepressants are all descendants of the serendipitous findings in the 1950s that the monoamine oxidase inhibitor iproniazid and the tricyclic antidepressant imipramine were effective antidepressants. Thus, the need for improved animal models to provide insights into the neuropathology underlying the disease is critical. Such information is in turn crucial for identifying new antidepressants and mood stabilisers. Currently, there is a shift away from traditional animal models to more focused research dealing with an endophenotype-style approach, genetic models, and incorporation of new findings from human neuroimaging and genetic studies. Such approaches are opening up more tractable avenues for understanding the neurobiological and genetic bases of these disorders. Further, such models promise to yield better translational animal models and hence more fruitful therapeutic targets. This overview focuses on such animal models and tests and how they can be used to assess MDD and BD in rodents.
Collapse
|
96
|
Llamosas N, Bruzos-Cidón C, Rodríguez JJ, Ugedo L, Torrecilla M. Deletion of GIRK2 Subunit of GIRK Channels Alters the 5-HT1A Receptor-Mediated Signaling and Results in a Depression-Resistant Behavior. Int J Neuropsychopharmacol 2015; 18:pyv051. [PMID: 25956878 PMCID: PMC4756724 DOI: 10.1093/ijnp/pyv051] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 05/04/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Targeting dorsal raphe 5-HT1A receptors, which are coupled to G-protein inwardly rectifying potassium (GIRK) channels, has revealed their contribution not only to behavioral and functional aspects of depression but also to the clinical response to its treatment. Although GIRK channels containing GIRK2 subunits play an important role controlling excitability of several brain areas, their impact on the dorsal raphe activity is still unknown. Thus, the goal of the present study was to investigate the involvement of GIRK2 subunit-containing GIRK channels in depression-related behaviors and physiology of serotonergic neurotransmission. METHODS Behavioral, functional, including in vivo extracellular recordings of dorsal raphe neurons, and neurogenesis studies were carried out in wild-type and GIRK2 mutant mice. RESULTS Deletion of the GIRK2 subunit promoted a depression-resistant phenotype and determined the behavioral response to the antidepressant citalopram without altering hippocampal neurogenesis. In dorsal raphe neurons of GIRK2 knockout mice, and also using GIRK channel blocker tertiapin-Q, the basal firing rate was higher than that obtained in wild-type animals, although no differences were observed in other firing parameters. 5-HT1A receptors were desensitized in GIRK2 knockout mice, as demonstrated by a lower sensitivity of dorsal raphe neurons to the inhibitory effect of the 5-HT1A receptor agonist, 8-OH-DPAT, and the antidepressant citalopram. CONCLUSIONS Our results indicate that GIRK channels formed by GIRK2 subunits determine depression-related behaviors as well as basal and 5-HT1A receptor-mediated dorsal raphe neuronal activity, becoming alternative therapeutic targets for psychiatric diseases underlying dysfunctional serotonin transmission.
Collapse
Affiliation(s)
| | | | | | | | - Maria Torrecilla
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU, Leioa, Spain (Drs Llamosas, Bruzos-Cidón, Ugedo, and Torrecilla); Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain (Dr Rodríguez); Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain (Dr Rodríguez).
| |
Collapse
|
97
|
Stepan J, Dine J, Eder M. Functional optical probing of the hippocampal trisynaptic circuit in vitro: network dynamics, filter properties, and polysynaptic induction of CA1 LTP. Front Neurosci 2015; 9:160. [PMID: 25999809 PMCID: PMC4422028 DOI: 10.3389/fnins.2015.00160] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 04/19/2015] [Indexed: 12/21/2022] Open
Abstract
Decades of brain research have identified various parallel loops linking the hippocampus with neocortical areas, enabling the acquisition of spatial and episodic memories. Especially the hippocampal trisynaptic circuit [entorhinal cortex layer II → dentate gyrus (DG) → cornu ammonis (CA)-3 → CA1] was studied in great detail because of its seemingly simple connectivity and characteristic structures that are experimentally well accessible. While numerous researchers focused on functional aspects, obtained from a limited number of cells in distinct hippocampal subregions, little is known about the neuronal network dynamics which drive information across multiple synapses for subsequent long-term storage. Fast voltage-sensitive dye imaging in vitro allows real-time recording of activity patterns in large/meso-scale neuronal networks with high spatial resolution. In this way, we recently found that entorhinal theta-frequency input to the DG most effectively passes filter mechanisms of the trisynaptic circuit network, generating activity waves which propagate across the entire DG-CA axis. These "trisynaptic circuit waves" involve high-frequency firing of CA3 pyramidal neurons, leading to a rapid induction of classical NMDA receptor-dependent long-term potentiation (LTP) at CA3-CA1 synapses (CA1 LTP). CA1 LTP has been substantially evidenced to be essential for some forms of explicit learning in mammals. Here, we review data with particular reference to whole network-level approaches, illustrating how activity propagation can take place within the trisynaptic circuit to drive formation of CA1 LTP.
Collapse
Affiliation(s)
- Jens Stepan
- Department Stress Neurobiology and Neurogenetics, Max Planck Institute of PsychiatryMunich, Germany
| | | | - Matthias Eder
- Department Stress Neurobiology and Neurogenetics, Max Planck Institute of PsychiatryMunich, Germany
| |
Collapse
|
98
|
McAvoy K, Russo C, Kim S, Rankin G, Sahay A. Fluoxetine induces input-specific hippocampal dendritic spine remodeling along the septotemporal axis in adulthood and middle age. Hippocampus 2015; 25:1429-46. [PMID: 25850664 DOI: 10.1002/hipo.22464] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2015] [Indexed: 12/15/2022]
Abstract
Fluoxetine, a selective serotonin-reuptake inhibitor (SSRI), is known to induce structural rearrangements and changes in synaptic transmission in hippocampal circuitry. In the adult hippocampus, structural changes include neurogenesis, dendritic, and axonal plasticity of pyramidal and dentate granule neurons, and dedifferentiation of dentate granule neurons. However, much less is known about how chronic fluoxetine affects these processes along the septotemporal axis and during the aging process. Importantly, studies documenting the effects of fluoxetine on density and distribution of spines along different dendritic segments of dentate granule neurons and CA1 pyramidal neurons along the septotemporal axis of hippocampus in adulthood and during aging are conspicuously absent. Here, we use a transgenic mouse line in which mature dentate granule neurons and CA1 pyramidal neurons are genetically labeled with green fluorescent protein (GFP) to investigate the effects of chronic fluoxetine treatment (18 mg/kg/day) on input-specific spine remodeling and mossy fiber structural plasticity in the dorsal and ventral hippocampus in adulthood and middle age. In addition, we examine levels of adult hippocampal neurogenesis, maturation state of dentate granule neurons, neuronal activity, and glutamic acid decarboxylase-67 expression in response to chronic fluoxetine in adulthood and middle age. Our studies reveal that while chronic fluoxetine fails to augment adult hippocampal neurogenesis in middle age, the middle-aged hippocampus retains high sensitivity to changes in the dentate gyrus (DG) such as dematuration, hypoactivation, and increased glutamic acid decarboxylase 67 (GAD67) expression. Interestingly, the middle-aged hippocampus shows greater sensitivity to fluoxetine-induced input-specific synaptic remodeling than the hippocampus in adulthood with the stratum-oriens of CA1 exhibiting heightened structural plasticity. The input-specific changes and circuit-level modifications in middle-age were associated with modest enhancement in contextual fear memory precision, anxiety-like behavior and antidepressant-like behavioral responses.
Collapse
Affiliation(s)
- Kathleen McAvoy
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Craig Russo
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shannen Kim
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Genelle Rankin
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
99
|
Abstract
Many patients with major depressive disorder (MDD) only partially respond, and some have no clinically meaningful response, to current widely used antidepressant drugs. Due to the purported role of dopamine in the pathophysiology of depression, triple-reuptake inhibitors (TRIs) that simultaneously inhibit serotonin (5-HT), norepinephrine (NE) and dopamine reuptake could be a useful addition to the armamentarium of treatments for MDD. A TRI should more effectively activate mesolimbic dopamine-related reward-networks, restore positive mood and reduce potent 5-HT reuptake blockade associated "hypodopaminergic" adverse effects of decreased libido, weight gain and "blunting" of emotions. On the other hand, dopaminergic effects raise concern over abuse liability and TRIs may have many of the cardiovascular effects associated with NET inhibition. Several clinical development programs for potential TRI antidepressants have failed to demonstrate significantly greater efficacy than placebo or standard of care. Successful late-stage clinical development of a TRI is more likely if experimental research studies in the target population of depressed patients have demonstrated target engagement that differentially and dose-dependently improves assessments of reward-network dysfunction relative to existing antidepressants. TRI treatment could be individualized on the basis of predictive markers such as the burden of decreased positive mood symptoms and/or neuroimaging evidence of reward network dysfunction. This review focuses on how the next generation of monoamine-based treatments could be efficiently developed to address unmet medical need in MDD.
Collapse
|
100
|
Li XL, Yuan YG, Xu H, Wu D, Gong WG, Geng LY, Wu FF, Tang H, Xu L, Zhang ZJ. Changed Synaptic Plasticity in Neural Circuits of Depressive-Like and Escitalopram-Treated Rats. Int J Neuropsychopharmacol 2015; 18:pyv046. [PMID: 25899067 PMCID: PMC4648155 DOI: 10.1093/ijnp/pyv046] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Although progress has been made in the detection and characterization of neural plasticity in depression, it has not been fully understood in individual synaptic changes in the neural circuits under chronic stress and antidepressant treatment. METHODS Using electron microscopy and Western-blot analyses, the present study quantitatively examined the changes in the Gray's Type I synaptic ultrastructures and the expression of synapse-associated proteins in the key brain regions of rats' depressive-related neural circuit after chronic unpredicted mild stress and/or escitalopram administration. Meanwhile, their depressive behaviors were also determined by several tests. RESULTS The Type I synapses underwent considerable remodeling after chronic unpredicted mild stress, which resulted in the changed width of the synaptic cleft, length of the active zone, postsynaptic density thickness, and/or synaptic curvature in the subregions of medial prefrontal cortex and hippocampus, as well as the basolateral amygdaloid nucleus of the amygdala, accompanied by changed expression of several synapse-associated proteins. Chronic escitalopram administration significantly changed the above alternations in the chronic unpredicted mild stress rats but had little effect on normal controls. Also, there was a positive correlation between the locomotor activity and the maximal synaptic postsynaptic density thickness in the stratum radiatum of the Cornu Ammonis 1 region and a negative correlation between the sucrose preference and the length of the active zone in the basolateral amygdaloid nucleus region in chronic unpredicted mild stress rats. CONCLUSION These findings strongly indicate that chronic stress and escitalopram can alter synaptic plasticity in the neural circuits, and the remodeled synaptic ultrastructure was correlated with the rats' depressive behaviors, suggesting a therapeutic target for further exploration.
Collapse
Affiliation(s)
- Xiao-Li Li
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Yong-Gui Yuan
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Hua Xu
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Di Wu
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Wei-Gang Gong
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Lei-Yu Geng
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Fang-Fang Wu
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Hao Tang
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Lin Xu
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Zhi-Jun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work.
| |
Collapse
|