1
|
Wang T, Gao C, Li J, Li L, Yue Y, Liu X, Chen S, Hou Z, Yin Y, Jiang W, Xu Z, Kong Y, Yuan Y. Prediction of Early Antidepressant Efficacy in Patients with Major Depressive Disorder Based on Multidimensional Features of rs-fMRI and P11 Gene DNA Methylation: Prédiction de l'efficacité précoce d'un antidépresseur chez des patients souffrant du trouble dépressif majeur d'après les caractéristiques multidimensionnelles de la méthylation de l'ADN du gène P11 et de la IRMf-rs. CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2024; 69:264-274. [PMID: 37920958 PMCID: PMC10924577 DOI: 10.1177/07067437231210787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
OBJECTIVE This study established a machine learning model based on the multidimensional data of resting-state functional activity of the brain and P11 gene DNA methylation to predict the early efficacy of antidepressant treatment in patients with major depressive disorder (MDD). METHODS A total of 98 Han Chinese MDD were analysed in this study. Patients were divided into 51 responders and 47 nonresponders according to whether the Hamilton Depression Rating Scale-17 items (HAMD-17) reduction rate was ≥50% after 2 weeks of antidepressant treatment. At baseline, the Illumina HiSeq Platform was used to detect the methylation of 74 CpG sites of the P11 gene in peripheral blood samples. Resting-state functional magnetic resonance imaging (rs-fMRI) scan detected the amplitude of low-frequency fluctuations (ALFF), regional homogeneity (ReHo), and functional connectivity (FC) in 116 brain regions. The least absolute shrinkage and selection operator analysis method was used to perform feature reduction and feature selection. Four typical machine learning methods were used to establish support vector machine (SVM), random forest (RF), Naïve Bayes (NB), and logistic regression (LR) prediction models based on different combinations of functional activity of the brain, P11 gene DNA methylation and clinical/demographic features after screening. RESULTS The SVM model based on ALFF, ReHo, FC, P11 methylation, and clinical/demographic features showed the best performance, with 95.92% predictive accuracy and 0.9967 area under the receiver operating characteristic curve, which was better than RF, NB, and LR models. CONCLUSION The multidimensional data features combining rs-fMRI, DNA methylation, and clinical/demographic features can predict the early antidepressant efficacy in MDD.
Collapse
Affiliation(s)
- Tianyu Wang
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Chenjie Gao
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jiaxing Li
- Jiangsu Provincial Joint International Research Laboratory of Medical Information Processing, School of Computer Science and Engineering, Southeast University, Nanjing, China
| | - Lei Li
- Department of Sleep Medicine, The Fourth People's Hospital of Lianyungang, Lianyungang, China
| | - Yingying Yue
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiaoyun Liu
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Suzhen Chen
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhenghua Hou
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yingying Yin
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Wenhao Jiang
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhi Xu
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Youyong Kong
- Jiangsu Provincial Joint International Research Laboratory of Medical Information Processing, School of Computer Science and Engineering, Southeast University, Nanjing, China
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Southeast University, Nanjing, China
| |
Collapse
|
2
|
Musaelyan K, Horowitz MA, McHugh S, Szele FG. Fluoxetine Can Cause Epileptogenesis and Aberrant Neurogenesis in Male Wild-Type Mice. Dev Neurosci 2023; 46:158-166. [PMID: 37302394 DOI: 10.1159/000531478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 05/05/2023] [Indexed: 06/13/2023] Open
Abstract
Antidepressants in general, and fluoxetine in particular, increase adult hippocampal neurogenesis (AHN) in mice. Here we asked how the antidepressant fluoxetine affects behavior and AHN in a corticosterone model of depression. In three groups of adult male C57BL/6j mice, we administered either vehicle (VEH), corticosterone (CORT) treatment to induce a depression-like state, or corticosterone plus a standard dose of fluoxetine (CORT+FLX). Following treatment, mice performed the open field test, the novelty suppressed feeding (NSF) test, and the splash test. Neurogenesis was assessed by means of immunohistochemistry using BrdU and neuronal maturation markers. Unexpectedly, 42% of the CORT+FLX-treated mice exhibited severe weight loss, seizures, and sudden death. As expected, the CORT-treated group had altered behaviors compared to the VEH group, but the CORT+FLX mice that survived did not show any behavioral improvement compared to the CORT group. Antidepressants generally increase neurogenesis and here we also found that compared to CORT mice, CORT+FLX mice that survived had a significantly greater density of BrdU+, BrdU+DCX+, and BrdU+NeuN+ cells, suggesting increased neurogenesis. Moreover, the density of BrdU+NeuN+ cells was increased in an aberrant location, the hilus, of CORT+FLX mice, similar to previous studies describing aberrant neurogenesis following seizures. In conclusion, fluoxetine could induce considerable adverse effects in wild-type mice, including seizure-like activity. Fluoxetine-induced neurogenesis increases could be related to this activity; therefore, proneurogenic effects of fluoxetine and other antidepressants, especially in the absence of any behavioral therapeutic effects, should be interpreted with caution.
Collapse
Affiliation(s)
- Ksenia Musaelyan
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Mark A Horowitz
- Research and Development Department, North East London NHS Foundation Trust, Ilford, UK
- Department of Psychiatry, University College London, London, UK
| | - Stephen McHugh
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Role of P11 through serotonergic and glutamatergic pathways in LID. Mol Biol Rep 2023; 50:4535-4549. [PMID: 36853472 DOI: 10.1007/s11033-023-08326-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 02/09/2023] [Indexed: 03/01/2023]
Abstract
Parkinson's disease is a progressive neurodegenerative disorder caused by the degeneration of dopaminergic neurons. This leads to the pathogenesis of multiple basal ganglia-thalamomotor loops and diverse neurotransmission alterations. Dopamine replacement therapy, and on top of that, levodopa and l-3,4-dihydroxyphenylalanine (L-DOPA), is the gold standard treatment, while it develops numerous complications. Levodopa-induced dyskinesia (LID) is well-known as the most prominent side effect. Several studies have been devoted to tackling this problem. Studies showed that metabotropic glutamate receptor 5 (mGluR5) antagonists and 5-hydroxytryptamine receptor 1B (5HT1B) agonists significantly reduced LID when considering the glutamatergic overactivity and compensatory mechanisms of serotonergic neurons after L-DOPA therapy. Moreover, it is documented that these receptors act through an adaptor protein called P11 (S100A10). This protein has been thought to play a crucial role in LID due to its interactions with numerous ion channels and receptors. Lately, experiments have shown successful evidence of the effects of P11 blockade on alleviating LID greater than 5HT1B and mGluR5 manipulations. In contrast, there is a trace of ambiguity in the exact mechanism of action. P11 has shown the potential to be a promising target to diminish LID and prolong L-DOPA therapy in parkinsonian patients owing to further studies and experiments.
Collapse
|
4
|
Larosa A, Wong TP. The hippocampus in stress susceptibility and resilience: Reviewing molecular and functional markers. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110601. [PMID: 35842073 DOI: 10.1016/j.pnpbp.2022.110601] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/22/2022] [Accepted: 07/10/2022] [Indexed: 10/17/2022]
Abstract
Understanding the individual variability that comes with the likelihood of developing stress-related psychopathologies is of paramount importance when addressing mechanisms of their neurobiology. This article focuses on the hippocampus as a region that is highly influenced by chronic stress exposure and that has strong ties to the development of related disorders, such as depression and post-traumatic stress disorder. We first outline three commonly used animal models that have been used to separate animals into susceptible and resilient cohorts. Next, we review molecular and functional hippocampal markers of susceptibility and resilience. We propose that the hippocampus plays a crucial role in the differences in the processing and storage of stress-related information in animals with different stress susceptibilities. These hippocampal markers not only help us attain a more comprehensive understanding of the various facets of stress-related pathophysiology, but also could be targeted for the development of new treatments.
Collapse
Affiliation(s)
- Amanda Larosa
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Tak Pan Wong
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada; Dept. of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
5
|
Wang T, Li L, Yue Y, Liu X, Chen S, Shen T, Xu Z, Yuan Y. The interaction of P11 methylation and early-life stress impacts the antidepressant response in patients with major depressive disorder. J Affect Disord 2022; 312:128-135. [PMID: 35752218 DOI: 10.1016/j.jad.2022.06.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/17/2022] [Accepted: 06/20/2022] [Indexed: 11/18/2022]
Abstract
PURPOSE The present research investigates the influence of P11 gene DNA methylation combined with life stress on the response to antidepressants in the first two weeks. METHODS A total of 291 Han Chinese patients with major depressive disorder and 100 healthy controls were included. The Life Events Scale and the Childhood Trauma Questionnaire were used to assess stress. The primary endpoint was the Hamilton Depression Rating Scale-17 reduction rate after two weeks of treatment. The Illumina HiSeq Platform was used to detect the methylation of 74 CpG sites of the P11 gene in peripheral blood samples. RESULTS The mean methylation of all P11 CpG sites, as well as the methylation at 4 CpG sites (P11-2-169, P11-2-192, P11-2-202, P11-2-204), were significantly higher in patients with MDD than in healthy controls (FDR-corrected P < 0.05). The response to antidepressants was associated with the following interactions: the CTQ score and P11-3-185 site methylation (OR = 0.297, FDR-corrected P = 0.023), the CTQ physical neglect score and P11-2-117 site methylation (OR = 0.005, FDR-corrected P = 0.033), and the CTQ emotional abuse score and P11-3-185 site methylation (OR = 0.001, FDR-corrected P = 0.023). CONCLUSIONS The methylation of the P11 gene was significantly higher in patients with major depressive disorder. The interaction of P11 DNA methylation and early-life stress may influence the short-term antidepressant treatment response.
Collapse
Affiliation(s)
- Tianyu Wang
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, PR China
| | - Lei Li
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, PR China; Department of Sleep Medicine, The Fourth People's Hospital of Lianyungang, Lianyungang 222000, PR China
| | - Yingying Yue
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, PR China
| | - Xiaoyun Liu
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, PR China
| | - Suzhen Chen
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, PR China
| | - Tian Shen
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, PR China
| | - Zhi Xu
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, PR China.
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, PR China; Jiangsu Provincial Key Laboratory of Critical Care Medicine, Southeast university, Nanjing 210009, PR China.
| |
Collapse
|
6
|
Brain serotonin deficiency and fluoxetine lead to sex-specific effects on binge-like food consumption in mice. Psychopharmacology (Berl) 2022; 239:2975-2984. [PMID: 35750862 DOI: 10.1007/s00213-022-06181-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/17/2022] [Indexed: 10/17/2022]
Abstract
RATIONALE Although pharmacotherapies are often effective in reducing binge eating in conditions such as bulimia nervosa and binge eating disorder, subsets of patients do not benefit sufficiently from existing treatments, and the reasons for treatment failure remain unclear. OBJECTIVES This study aimed to evaluate whether genetic reductions in brain serotonin influence binge eating and/or the ability of fluoxetine, a selective serotonin reuptake inhibitor, to reduce binge eating in mice. METHODS This study used a validated model of binge-like consumption of high-fat diet to compare binge-like food intake in control and fluoxetine-treated wild-type and serotonin-deficient mice from the tryptophan hydroxylase 2 (R439H) knock-in line. In addition, real-time PCR was used to evaluate potential genotype and sex differences in the effects of fluoxetine on gene expression in the raphe nucleus. RESULTS The results reveal that brain serotonin deficiency is sufficient to increase binge eating in males, but not females. However, while chronic fluoxetine reduced binge eating in both genotypes of males and in wild-type females, it failed to reduce binge eating in serotonin-deficient females. Transcriptional responses to chronic fluoxetine were also characterized by sex and genotype differences. CONCLUSIONS Overall, this study revealed significant sex differences in the effects of fluoxetine and brain serotonin deficiency on binge-like food intake and suggests that low brain serotonin could impact eating disorders both by promoting binge eating and by limiting the efficacy of fluoxetine to reduce binge eating.
Collapse
|
7
|
Chen MX, Oh YS, Kim Y. S100A10 and its binding partners in depression and antidepressant actions. Front Mol Neurosci 2022; 15:953066. [PMID: 36046712 PMCID: PMC9423026 DOI: 10.3389/fnmol.2022.953066] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
S100A10 (p11) is an emerging player in the neurobiology of depression and antidepressant actions. p11 was initially thought to be a modulator of serotonin receptor (5-HTR) trafficking and serotonergic transmission, though newly identified binding partners of p11 and neurobiological studies of these proteins have shed light on multifunctional roles for p11 in the regulation of glutamatergic transmission, calcium signaling and nuclear events related to chromatin remodeling, histone modification, and gene transcription. This review article focuses on direct binding partners of p11 in the brain including 5-HTRs, mGluR5, annexin A2, Ahnak, Smarca3, and Supt6h, as well as their roles in neuronal function, particularly in the context of depressive-like behavior as well as behavioral effects of antidepressant drug treatments in mice. In addition, we discuss neurobiological insights from recently uncovered p11 pathways in multiple types of neurons and non-neuronal cells and cast major remaining questions for future studies.
Collapse
Affiliation(s)
- Michelle X. Chen
- University of Iowa Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa, IA, United States
| | - Yong-Seok Oh
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Daegu, South Korea
| | - Yong Kim
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States
- Brain Health Institute, Rutgers University, Piscataway, NJ, United States
- *Correspondence: Yong Kim
| |
Collapse
|
8
|
Ling F, Lu Q. S100 calcium-binding protein A10 contributes to malignant traits in osteosarcoma cells by regulating glycolytic metabolism via the AKT/mTOR pathway. Bioengineered 2022; 13:12298-12308. [PMID: 35579448 PMCID: PMC9276053 DOI: 10.1080/21655979.2022.2071022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
As an aggressive musculoskeletal malignancy, osteosarcoma (OSa) is popular among young adults and teenagers worldwide. S100 calcium-binding protein A10 (S100A10) functioned as a novel tumor-promoting protein in several human cancers. However, its role in OSa remains obscure. In this study, gene and protein levels were respectively determined by RT-qPCR or Western blotting. OSa cell proliferation, apoptosis, and metastasis were evaluated via CCK-8, colony formation, flow cytometry, and Transwell assays. To assess the glycolysis level, glucose consumption and lactate production were detected. It was found S100A10 was highly expressed in OSa tissues and cell lines. Besides, S100A10 facilitated proliferation and metastasis, and inhibited apoptosis in OSa cells. In addition, S100A10 regulated OSa cell proliferation, metastasis and apoptosis via mediating the glycolysis process. Furthermore, S100A10-mediated AKT/mTOR signaling accelerated glycolysis, thereby promoting malignant behaviors in OSa cells. Taken together, our findings indicated that S100A10 might promote malignant phenotypes of OSa cells by accelerating glycolysis and activating the AKT/mTOR signaling, providing a promising target for OSa treatment.
Collapse
Affiliation(s)
- Feng Ling
- Department of Trauma Orthopaedics, Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Qifeng Lu
- Department of Trauma Orthopaedics, Taizhou People's Hospital, Taizhou, Jiangsu, China
| |
Collapse
|
9
|
Seo MK, Lee JG, Park SW. Early life stress induces age-dependent epigenetic changes in p11 gene expression in male mice. Sci Rep 2021; 11:10663. [PMID: 34471143 PMCID: PMC8410943 DOI: 10.1038/s41598-021-89593-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
Early life stress (ELS) causes long-lasting changes in gene expression through epigenetic mechanisms. However, little is known about the effects of ELS in adulthood, specifically across different age groups. In this study, the epigenetic modifications of p11 expression in adult mice subjected to ELS were investigated in different stages of adulthood. Pups experienced maternal separation (MS) for 3 h daily from postnatal day 1 to 21. At young and middle adulthood, behavioral test, hippocampal p11 expression levels, and levels of histone acetylation and methylation and DNA methylation at the hippocampal p11 promoter were measured. Middle-aged, but not young adult, MS mice exhibited increased immobility time in the forced swimming test. Concurrent with reduced hippocampal p11 levels, mice in both age groups showed a decrease in histone acetylation (AcH3) and permissive histone methylation (H3K4me3) at the p11 promoter, as well as an increase in repressive histone methylation (H3K27me3). Moreover, our results showed that the expression, AcH3 and H3Kme3 levels of p11 gene in response to MS were reduced with age. DNA methylation analysis of the p11 promoter revealed increased CpG methylation in middle-aged MS mice only. The results highlight the age-dependent deleterious effects of ELS on the epigenetic modifications of p11 transcription.
Collapse
Affiliation(s)
- Mi Kyoung Seo
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea
| | - Jung Goo Lee
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea. .,Department of Psychiatry, College of Medicine, Haeundae Paik Hospital, Inje University, Busan, 48108, Republic of Korea.
| | - Sung Woo Park
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea. .,Department of Convergence Biomedical Science, College of Medicine, Inje University, Busan, 47392, Republic of Korea.
| |
Collapse
|
10
|
Umschweif G, Medrihan L, McCabe KA, Sagi Y, Greengard P. Activation of the p11/SMARCA3/Neurensin-2 pathway in parvalbumin interneurons mediates the response to chronic antidepressants. Mol Psychiatry 2021; 26:3350-3362. [PMID: 33723417 PMCID: PMC8505248 DOI: 10.1038/s41380-021-01059-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 02/02/2021] [Accepted: 02/18/2021] [Indexed: 12/26/2022]
Abstract
The delayed behavioral response to chronic antidepressants depends on dynamic changes in the hippocampus. It was suggested that the antidepressant protein p11 and the chromatin remodeling factor SMARCA3 mediate this delayed response by inducing transcriptional changes in hippocampal neurons. However, what target genes are regulated by the p11/SMARCA3 complex to mediate the behavioral response to antidepressants, and what cell type mediates these molecular changes remain unknown. Here we report that the p11/SMARCA3 complex represses Neurensin-2 transcription in hippocampal parvalbumin-expressing interneurons after chronic treatment with Selective Serotonin Reuptake Inhibitors (SSRI). The behavioral response to antidepressants requires upregulation of p11, accumulation of SMARCA3 in the cell nucleus, and a consequent repression of Neurensin-2 transcription in these interneurons. We elucidate a functional role for p11/SMARCA3/Neurensin-2 pathway in regulating AMPA-receptor signaling in parvalbumin-expressing interneurons, a function that is enhanced by chronic treatment with SSRIs. These results link SSRIs to dynamic glutamatergic changes and implicate p11/SMARCA3/Neurensin-2 pathway in the development of more specific and efficient therapeutic strategies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Gali Umschweif
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| | - Lucian Medrihan
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| | - Kathryn A McCabe
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| | - Yotam Sagi
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA.
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| |
Collapse
|
11
|
Umschweif G, Medrihan L, Guillén-Samander A, Wang W, Sagi Y, Greengard P. Identification of Neurensin-2 as a novel modulator of emotional behavior. Mol Psychiatry 2021; 26:2872-2885. [PMID: 33742167 PMCID: PMC8505262 DOI: 10.1038/s41380-021-01058-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 02/02/2021] [Accepted: 02/18/2021] [Indexed: 11/09/2022]
Abstract
Among the hallmarks of major depressive disorders (MDD) are molecular, functional, and morphological impairments in the hippocampus. Recent studies suggested a key role for hippocampal GABAergic interneurons both in depression and in the response to its treatments. These interneurons highly express the chromatin-remodeler SMARCA3 which mediates the response to chronic antidepressants in an unknown mechanism. Using cell-type-specific molecular and physiological approaches, we report that SMARCA3 mediates the glutamatergic signaling in interneurons by repressing the expression of the neuronal protein, Neurensin-2. This vesicular protein associates with endosomes and postsynaptic proteins and is highly and selectively expressed in subpopulations of GABAergic interneurons. Upregulation of Neurensin-2 in the hippocampus either by stress, viral overexpression, or by SMARCA3 deletion, results in depressive-like behaviors. In contrast, the deletion of Neurensin-2 confers resilience to stress and induces AMPA receptor localization to synapses. This pathway which bidirectionally affects emotional behavior could be involved in neuropsychiatric disorders, and suggests novel therapeutic approaches.
Collapse
Affiliation(s)
- Gali Umschweif
- Laboratory for Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| | - Lucian Medrihan
- Laboratory for Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| | | | - Wei Wang
- Laboratory for Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| | - Yotam Sagi
- Laboratory for Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA.
| | - Paul Greengard
- Laboratory for Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| |
Collapse
|
12
|
Role of adult hippocampal neurogenesis in the antidepressant actions of lactate. Mol Psychiatry 2021; 26:6723-6735. [PMID: 33990772 PMCID: PMC8760055 DOI: 10.1038/s41380-021-01122-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 02/03/2023]
Abstract
In addition to its role as a neuronal energy substrate and signaling molecule involved in synaptic plasticity and memory consolidation, recent evidence shows that lactate produces antidepressant effects in animal models. However, the mechanisms underpinning lactate's antidepressant actions remain largely unknown. In this study, we report that lactate reverses the effects of corticosterone on depressive-like behavior, as well as on the inhibition of both the survival and proliferation of new neurons in the adult hippocampus. Furthermore, the inhibition of adult hippocampal neurogenesis prevents the antidepressant-like effects of lactate. Pyruvate, the oxidized form of lactate, did not mimic the effects of lactate on adult hippocampal neurogenesis and depression-like behavior. Finally, our data suggest that conversion of lactate to pyruvate with the concomitant production of NADH is necessary for the neurogenic and antidepressant effects of lactate.
Collapse
|
13
|
P11 deficiency increases stress reactivity along with HPA axis and autonomic hyperresponsiveness. Mol Psychiatry 2021; 26:3253-3265. [PMID: 33005029 PMCID: PMC8505237 DOI: 10.1038/s41380-020-00887-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/02/2020] [Accepted: 09/11/2020] [Indexed: 12/22/2022]
Abstract
Patients suffering from mood disorders and anxiety commonly exhibit hypothalamic-pituitary-adrenocortical (HPA) axis and autonomic hyperresponsiveness. A wealth of data using preclinical animal models and human patient samples indicate that p11 deficiency is implicated in depression-like phenotypes. In the present study, we used p11-deficient (p11KO) mice to study potential roles of p11 in stress responsiveness. We measured stress response using behavioral, endocrine, and physiological readouts across early postnatal and adult life. Our data show that p11KO pups respond more strongly to maternal separation than wild-type pups, even though their mothers show no deficits in maternal behavior. Adult p11KO mice display hyperactivity of the HPA axis, which is paralleled by depression- and anxiety-like behaviors. p11 was found to be highly enriched in vasopressinergic cells of the paraventricular nucleus and regulates HPA hyperactivity in a V1B receptor-dependent manner. Moreover, p11KO mice display sympathetic-adrenal-medullary (SAM) axis hyperactivity, with elevated adrenal norepinephrine and epinephrine levels. Using conditional p11KO mice, we demonstrate that this SAM hyperactivity is partially regulated by the loss of p11 in serotonergic neurons of the raphe nuclei. Telemetric electrocardiogram measurements show delayed heart rate recovery in p11KO mice in response to novelty exposure and during expression of fear following auditory trace fear conditioning. Furthermore, p11KO mice have elevated basal heart rate in fear conditioning tests indicating increased autonomic responsiveness. This set of experiments provide strong and versatile evidence that p11 deficiency leads to HPA and SAM axes hyperresponsiveness along with increased stress reactivity.
Collapse
|
14
|
Medrihan L, Umschweif G, Sinha A, Reed S, Lee J, Gindinova K, Sinha SC, Greengard P, Sagi Y. Reduced Kv3.1 Activity in Dentate Gyrus Parvalbumin Cells Induces Vulnerability to Depression. Biol Psychiatry 2020; 88:405-414. [PMID: 32331822 DOI: 10.1016/j.biopsych.2020.02.1179] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 01/30/2020] [Accepted: 02/18/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Parvalbumin (PV)-expressing interneurons are important for cognitive and emotional behaviors. These neurons express high levels of p11, a protein associated with depression and action of antidepressants. METHODS We characterized the behavioral response to subthreshold stress in mice with conditional deletion of p11 in PV cells. Using chemogenetics, viral-mediated gene delivery, and a specific ion channel agonist, we studied the role of dentate gyrus PV cells in regulating anxiety-like behavior and resilience to stress. We used electrophysiology, imaging, and biochemical studies in mice and cells to elucidate the function and mechanism of p11 in dentate gyrus PV cells. RESULTS p11 regulates the subcellular localization and cellular level of the potassium channel Kv3.1 in cells. Deletion of p11 from PV cells resulted in reduced hippocampal level of Kv3.1, attenuated capacity of high-frequency firing in dentate gyrus PV cells, and altered short-term plasticity at synapses on granule cells, as well as anxiety-like behavior and a pattern separation deficit. Chemogenetic inhibition or deletion of p11 in these cells induced vulnerability to depressive behavior, whereas upregulation of Kv3.1 in dentate gyrus PV cells or acute activation of Kv3.1 using a specific agonist induced resilience to depression. CONCLUSIONS The activity of dentate gyrus PV cells plays a major role in the behavioral response to novelty and stress. Activation of the Kv3.1 channel in dentate gyrus PV cells may represent a target for the development of cell-type specific, fast-acting antidepressants.
Collapse
Affiliation(s)
- Lucian Medrihan
- Laboratory for Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Gali Umschweif
- Laboratory for Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Anjana Sinha
- Laboratory for Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Shayna Reed
- Laboratory for Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Jinah Lee
- Laboratory for Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Katherina Gindinova
- Laboratory for Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Subhash C Sinha
- Laboratory for Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Paul Greengard
- Laboratory for Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Yotam Sagi
- Laboratory for Molecular and Cellular Neuroscience, Rockefeller University, New York, New York.
| |
Collapse
|
15
|
Systemic immunization with altered myelin basic protein peptide produces sustained antidepressant-like effects. Mol Psychiatry 2020; 25:1260-1274. [PMID: 31375779 DOI: 10.1038/s41380-019-0470-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 06/17/2019] [Accepted: 06/24/2019] [Indexed: 12/21/2022]
Abstract
Immune dysregulation, specifically of inflammatory processes, has been linked to behavioral symptoms of depression in both human and rodent studies. Here, we evaluated the antidepressant effects of immunization with altered peptide ligands of myelin basic protein (MBP)-MBP87-99[A91, A96], MBP87-99[A91], and MBP87-99[R91, A96]-in different models of depression and examined the mechanism by which these peptides protect against stress-induced depression. We found that a single dose of subcutaneously administered MBP87-99[A91, A96] produced antidepressant-like effects by decreasing immobility in the forced swim test and by reducing the escape latency and escape failures in the learned helplessness paradigm. Moreover, immunization with MBP87-99[A91, A96] prevented and reversed depressive-like and anxiety-like behaviors that were induced by chronic unpredictable stress (CUS). However, MBP87-99[R91, A96] tended to aggravate CUS-induced anxiety-like behavior. Chronic stress increased the production of peripheral and central proinflammatory cytokines and induced the activation of microglia in the prelimbic cortex (PrL), which was blocked by MBP87-99[A91, A96]. Immunization with MBP-derived altered peptide ligands also rescued chronic stress-induced deficits in p11, phosphorylated cyclic adenosine monophosphate response element binding protein, and brain-derived neurotrophic factor expression. Moreover, microinjections of recombinant proinflammatory cytokines and the knockdown of p11 in the PrL blunted the antidepressant-like behavioral response to MBP87-99[A91, A96]. Altogether, these findings indicate that immunization with altered MBP peptide produces prolonged antidepressant-like effects in rats, and the behavioral response is mediated by inflammatory factors (particularly interleukin-6), and p11 signaling in the PrL. Immune-neural interactions may impact central nervous system function and alter an individual's response to stress.
Collapse
|
16
|
Oh SJ, Cheng J, Jang JH, Arace J, Jeong M, Shin CH, Park J, Jin J, Greengard P, Oh YS. Hippocampal mossy cell involvement in behavioral and neurogenic responses to chronic antidepressant treatment. Mol Psychiatry 2020; 25:1215-1228. [PMID: 30837688 DOI: 10.1038/s41380-019-0384-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 01/29/2019] [Accepted: 02/14/2019] [Indexed: 12/28/2022]
Abstract
Most antidepressants, including selective serotonin reuptake inhibitors (SSRIs), initiate their drug actions by rapid elevation of serotonin, but they take several weeks to achieve therapeutic onset. This therapeutic delay suggests slow adaptive changes in multiple neuronal subtypes and their neural circuits over prolonged periods of drug treatment. Mossy cells are excitatory neurons in the dentate hilus that regulate dentate gyrus activity and function. Here we show that neuronal activity of hippocampal mossy cells is enhanced by chronic, but not acute, SSRI administration. Behavioral and neurogenic effects of chronic treatment with the SSRI, fluoxetine, are abolished by mossy cell-specific knockout of p11 or Smarca3 or by an inhibition of the p11/AnxA2/SMARCA3 heterohexamer, an SSRI-inducible protein complex. Furthermore, simple chemogenetic activation of mossy cells using Gq-DREADD is sufficient to elevate the proliferation and survival of the neural stem cells. Conversely, acute chemogenetic inhibition of mossy cells using Gi-DREADD impairs behavioral and neurogenic responses to chronic administration of SSRI. The present data establish that mossy cells play a crucial role in mediating the effects of chronic antidepressant medication. Our results indicate that compounds that target mossy cell activity would be attractive candidates for the development of new antidepressant medications.
Collapse
Affiliation(s)
- Seo-Jin Oh
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Hyenpung-myeon, Dalseong-gun, Daegu, Republic of Korea
| | - Jia Cheng
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, 10065, USA
| | - Jin-Hyeok Jang
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Hyenpung-myeon, Dalseong-gun, Daegu, Republic of Korea
| | - Jeffrey Arace
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, 10065, USA
| | - Minseok Jeong
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Hyenpung-myeon, Dalseong-gun, Daegu, Republic of Korea
| | - Chang-Hoon Shin
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Hyenpung-myeon, Dalseong-gun, Daegu, Republic of Korea
| | - Jeongrak Park
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Hyenpung-myeon, Dalseong-gun, Daegu, Republic of Korea
| | - Junghee Jin
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, 10065, USA
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, 10065, USA
| | - Yong-Seok Oh
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Hyenpung-myeon, Dalseong-gun, Daegu, Republic of Korea.
| |
Collapse
|
17
|
Neuroprotective efficacy of different levels of high-frequency repetitive transcranial magnetic stimulation in mice with CUMS-induced depression: Involvement of the p11/BDNF/Homer1a signaling pathway. J Psychiatr Res 2020; 125:152-163. [PMID: 32289652 DOI: 10.1016/j.jpsychires.2020.03.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 03/03/2020] [Accepted: 03/31/2020] [Indexed: 12/18/2022]
Abstract
High-frequency repetitive transcranial magnetic stimulation (HF-rTMS) is widely used to treat depression. However, the underlying mechanism has not been identified, and there is uncertainty regarding the optimal choice of stimulus parameters, especially stimulus frequency. Our previous study in mice demonstrated that 10-Hz HF-rTMS ameliorated depression by inducing expression of Homer1a and reducing excitability of cortical pyramidal cells. The aims of this study were to compare the effects of 15-Hz and 25-Hz HF-rTMS in a model of chronic unpredictable mild stress (CUMS)-induced depression and investigate its possible molecular mechanism. Male C57BL/6J mice were treated with CUMS for 28 days followed by 15-Hz and 25-Hz rTMS for 4 weeks. The sucrose preference, open field, forced swimming, and tail suspension tests were used to evaluate depression-like behaviors. Immunostaining was performed to measure neuronal loss and neurogenesis. Apoptosis was detected by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining. Expression of synapse-related proteins and the effects of HF-rTMS on the signaling pathway were examined using Western blot. The results showed that both 15-Hz and 25-Hz rTMS had significant antidepressant effects; 15-Hz rTMS seemed to be more effective than 25-Hz rTMS in preventing neuronal loss and promoting neurogenesis, while 25-Hz rTMS was superior to 15-Hz rTMS in facilitating synaptic plasticity. We also found that 15-Hz and 25-Hz rTMS markedly increased expression of p11, BDNF, Homer1a, and p-trkB proteins. These findings suggest that 15-Hz and 25-Hz HF-rTMS could exert neuroprotective effects to different degrees via multiple perspectives, which at least in part involve the p11/BDNF/Homer1a pathway.
Collapse
|
18
|
Jin J, Bhatti DL, Lee KW, Medrihan L, Cheng J, Wei J, Zhong P, Yan Z, Kooiker C, Song C, Ahn JH, Obermair GJ, Lee A, Gresack J, Greengard P, Kim Y. Ahnak scaffolds p11/Anxa2 complex and L-type voltage-gated calcium channel and modulates depressive behavior. Mol Psychiatry 2020; 25:1035-1049. [PMID: 30760886 PMCID: PMC6692256 DOI: 10.1038/s41380-019-0371-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 12/14/2018] [Accepted: 01/11/2019] [Indexed: 01/05/2023]
Abstract
Genetic polymorphisms of the L-type voltage-gated calcium channel (VGCC) are associated with psychiatric disorders including major depressive disorder. Alterations of S100A10 (p11) level are also implicated in the etiology of major depressive disorder. However, the existence of an endogenous regulator in the brain regulating p11, L-type VGCC, and depressive behavior has not been known. Here we report that Ahnak, whose function in the brain has been obscure, stabilizes p11 and Anxa2 proteins in the hippocampus and prefrontal cortex in the rodent brain. Protein levels of Ahnak, p11, and Anxa2 are highly and positively correlated in the brain. Together these data suggest the existence of an Ahnak/p11/Anxa2 protein complex. Ahnak is expressed in p11-positive as well as p11-negative neurons. Ahnak, through its N-terminal region, scaffolds the L-type pore-forming α1 subunit and, through its C-terminal region, scaffolds the β subunit of VGCC and the p11/Anxa2 complex. Cell surface expression of the α1 subunits and L-type calcium current are significantly reduced in primary cultures of Ahnak knockout (KO) neurons compared to wild-type controls. A decrease in the L-type calcium influx is observed in both glutamatergic neurons and parvalbumin (PV) GABAergic interneurons of Ahnak KO mice. Constitutive Ahnak KO mice or forebrain glutamatergic neuron-selective Ahnak KO mice display a depression-like behavioral phenotype similar to that of constitutive p11 KO mice. In contrast, PV interneuron-selective Ahnak KO mice display an antidepressant-like behavioral phenotype. Our results demonstrate L-type VGCC as an effector of the Ahnak/p11/Anxa2 complex, revealing a novel molecular connection involved in the control of depressive behavior.
Collapse
Affiliation(s)
- Junghee Jin
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Dionnet L. Bhatti
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Ko-Woon Lee
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Lucian Medrihan
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Jia Cheng
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Jing Wei
- 0000 0004 1936 9887grid.273335.3Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY USA
| | - Ping Zhong
- 0000 0004 1936 9887grid.273335.3Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY USA
| | - Zhen Yan
- 0000 0004 1936 9887grid.273335.3Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY USA
| | - Cassandra Kooiker
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Claire Song
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Jung-Hyuck Ahn
- 0000 0001 2171 7754grid.255649.9Department of Biochemistry, Ewha Womans University, Seoul, South Korea
| | - Gerald J. Obermair
- 0000 0000 8853 2677grid.5361.1Division of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Amy Lee
- 0000 0004 1936 8294grid.214572.7Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA USA
| | - Jodi Gresack
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Paul Greengard
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Yong Kim
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
19
|
Sargin D, Chottekalapanda RU, Perit KE, Yao V, Chu D, Sparks DW, Kalik S, Power SK, Troyanskaya OG, Schmidt EF, Greengard P, Lambe EK. Mapping the physiological and molecular markers of stress and SSRI antidepressant treatment in S100a10 corticostriatal neurons. Mol Psychiatry 2020; 25:1112-1129. [PMID: 31431686 PMCID: PMC7031043 DOI: 10.1038/s41380-019-0473-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 04/08/2019] [Accepted: 05/17/2019] [Indexed: 12/01/2022]
Abstract
In mood disorders, psychomotor and sensory abnormalities are prevalent, disabling, and intertwined with emotional and cognitive symptoms. Corticostriatal neurons in motor and somatosensory cortex are implicated in these symptoms, yet mechanisms of their vulnerability are unknown. Here, we demonstrate that S100a10 corticostriatal neurons exhibit distinct serotonin responses and have increased excitability, compared with S100a10-negative neurons. We reveal that prolonged social isolation disrupts the specific serotonin response which gets restored by chronic antidepressant treatment. We identify cell-type-specific transcriptional signatures in S100a10 neurons that contribute to serotonin responses and strongly associate with psychomotor and somatosensory function. Our studies provide a strong framework to understand the pathogenesis and create new avenues for the treatment of mood disorders.
Collapse
Affiliation(s)
- Derya Sargin
- 0000 0001 2157 2938grid.17063.33Department of Physiology, University of Toronto, Toronto, ON Canada
| | - Revathy U. Chottekalapanda
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065 USA
| | - Kristina E. Perit
- 0000 0001 2157 2938grid.17063.33Department of Physiology, University of Toronto, Toronto, ON Canada
| | - Victoria Yao
- 0000 0001 2097 5006grid.16750.35Department of Computer Science, Princeton University, Princeton, NJ 08544 USA ,0000 0001 2097 5006grid.16750.35Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544 USA
| | - Duong Chu
- 0000 0001 2157 2938grid.17063.33Department of Physiology, University of Toronto, Toronto, ON Canada
| | - Daniel W. Sparks
- 0000 0001 2157 2938grid.17063.33Department of Physiology, University of Toronto, Toronto, ON Canada
| | - Salina Kalik
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065 USA
| | - Saige K. Power
- 0000 0001 2157 2938grid.17063.33Department of Physiology, University of Toronto, Toronto, ON Canada
| | - Olga G. Troyanskaya
- 0000 0001 2097 5006grid.16750.35Department of Computer Science, Princeton University, Princeton, NJ 08544 USA ,0000 0001 2097 5006grid.16750.35Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544 USA ,grid.430264.7Flatiron Institute, Simons Foundation, New York, NY 10010 USA
| | - Eric F. Schmidt
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular Biology, The Rockefeller University, New York, NY 10065 USA
| | - Paul Greengard
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065 USA
| | - Evelyn K. Lambe
- 0000 0001 2157 2938grid.17063.33Department of Physiology, University of Toronto, Toronto, ON Canada ,0000 0001 2157 2938grid.17063.33Department of OBGYN, University of Toronto, Toronto, ON Canada ,0000 0001 2157 2938grid.17063.33Department of Psychiatry, University of Toronto, Toronto, ON Canada
| |
Collapse
|
20
|
Chottekalapanda RU, Kalik S, Gresack J, Ayala A, Gao M, Wang W, Meller S, Aly A, Schaefer A, Greengard P. AP-1 controls the p11-dependent antidepressant response. Mol Psychiatry 2020; 25:1364-1381. [PMID: 32439846 PMCID: PMC7303013 DOI: 10.1038/s41380-020-0767-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 04/10/2020] [Accepted: 04/28/2020] [Indexed: 01/10/2023]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the most widely prescribed drugs for mood disorders. While the mechanism of SSRI action is still unknown, SSRIs are thought to exert therapeutic effects by elevating extracellular serotonin levels in the brain, and remodel the structural and functional alterations dysregulated during depression. To determine their precise mode of action, we tested whether such neuroadaptive processes are modulated by regulation of specific gene expression programs. Here we identify a transcriptional program regulated by activator protein-1 (AP-1) complex, formed by c-Fos and c-Jun that is selectively activated prior to the onset of the chronic SSRI response. The AP-1 transcriptional program modulates the expression of key neuronal remodeling genes, including S100a10 (p11), linking neuronal plasticity to the antidepressant response. We find that AP-1 function is required for the antidepressant effect in vivo. Furthermore, we demonstrate how neurochemical pathways of BDNF and FGF2, through the MAPK, PI3K, and JNK cascades, regulate AP-1 function to mediate the beneficial effects of the antidepressant response. Here we put forth a sequential molecular network to track the antidepressant response and provide a new avenue that could be used to accelerate or potentiate antidepressant responses by triggering neuroplasticity.
Collapse
Affiliation(s)
- Revathy U. Chottekalapanda
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Salina Kalik
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Jodi Gresack
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Alyssa Ayala
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Melanie Gao
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Wei Wang
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Sarah Meller
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Ammar Aly
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Anne Schaefer
- 0000 0001 0670 2351grid.59734.3cFriedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Paul Greengard
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| |
Collapse
|
21
|
PSA-NCAM Colocalized with Cholecystokinin-Expressing Cells in the Hippocampus Is Involved in Mediating Antidepressant Efficacy. J Neurosci 2019; 40:825-842. [PMID: 31801810 DOI: 10.1523/jneurosci.1779-19.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/30/2019] [Accepted: 11/25/2019] [Indexed: 11/21/2022] Open
Abstract
The extracellular glycan polysialic acid linked to neural cell adhesion molecule (PSA-NCAM) is principally expressed in the developing brain and the adult neurogenic regions. Although colocalization of PSA-NCAM with cholecystokinin (CCK) was found in the adult brain, the role of PSA-NCAM remains unclear. In this study, we aimed to elucidate the functional significance of PSA-NCAM in the CA1 region of the male mouse hippocampus. Combined fluorescence in situ hybridization and immunohistochemistry showed that few vesicular glutamate transporter 3-negative/CCK-positive (VGluT3-/CCK+) cells were colocalized with PSA-NCAM, but most of the VGluT3+/CCK+ cells were colocalized with PSA-NCAM. The somata of PSA-NCAM+/CCK+ cells were highly innervated by serotonergic boutons than those of PSA-NCAM-/CCK+ cells. The expression ratios of 5-HT3A receptors and p11, a serotonin receptor-interacting protein, were higher in PSA-NCAM+/CCK+ cells than in PSA-NCAM-/CCK+ cells. Pharmacological digestion of PSA-NCAM impaired the efficacy of antidepressant fluoxetine (FLX), a selective serotonin reuptake inhibitor, but not the efficacy of benzodiazepine anxiolytic diazepam. A Western blot showed that restraint stress decreased the expressions of p11 and mature brain-derived neurotrophic factor (BDNF), and FLX increased them. Interestingly, the FLX-induced elevation of expression of p11, but not mature BDNF, was impaired by the digestion of PSA-NCAM. Quantitative reverse transcription-polymerase chain reaction showed that restraint stress reduced the expression of polysialyltransferase ST8Sia IV and FLX elevated it. Collectively, PSA-NCAM colocalized with VGluT3+/CCK+ cells in the CA1 region of the hippocampus may play a unique role in the regulation of antidepressant efficacy via the serotonergic pathway.SIGNIFICANCE STATEMENT Polysialic acid (PSA) is composed of eight or more α2,8-linked sialic acids. Here, we examined the functional significance of polysialic acid linked to the neural cell adhesion molecule (PSA-NCAM) in the adult mouse hippocampus. Few vesicular glutamate transporter 3-negative/cholecystokinin-positive (VGluT3-/CCK+) cells were colocalized with PSA-NCAM, but most of the VGluT3+/CCK+ cells were colocalized with PSA-NCAM. The expression ratios of 5-HT3A receptors and p11, a serotonin receptor-interacting protein, were higher in PSA-NCAM+/CCK+ cells than in PSA-NCAM-/CCK+ cells. The efficacy of antidepressants, but not anxiolytics, was impaired by the digestion of PSA-NCAM. The antidepressant-induced increase in p11 expression was inhibited following PSA-NCAM digestion. We hence hypothesize that PSA-NCAM colocalized with VGluT3+/CCK+ cells may play a unique role in regulating antidepressant efficacy.
Collapse
|
22
|
Planchez B, Surget A, Belzung C. Animal models of major depression: drawbacks and challenges. J Neural Transm (Vienna) 2019; 126:1383-1408. [PMID: 31584111 PMCID: PMC6815270 DOI: 10.1007/s00702-019-02084-y] [Citation(s) in RCA: 238] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022]
Abstract
Major depression is a leading contributor to the global burden of disease. This situation is mainly related to the chronicity and/or recurrence of the disorder, and to poor response to antidepressant therapy. Progress in this area requires valid animal models. Current models are based either on manipulating the environment to which rodents are exposed (during the developmental period or adulthood) or biological underpinnings (i.e. gene deletion or overexpression of candidate genes, targeted lesions of brain areas, optogenetic control of specific neuronal populations, etc.). These manipulations can alter specific behavioural and biological outcomes that can be related to different symptomatic and pathophysiological dimensions of major depression. However, animal models of major depression display substantial shortcomings that contribute to the lack of innovative pharmacological approaches in recent decades and which hamper our capabilities to investigate treatment-resistant depression. Here, we discuss the validity of these models, review putative models of treatment-resistant depression, major depression subtypes and recurrent depression. Furthermore, we identify future challenges regarding new paradigms such as those proposing dimensional rather than categorical approaches to depression.
Collapse
Affiliation(s)
| | | | - Catherine Belzung
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
- UMR 1253, iBrain, UFR Sciences et Techniques, Parc Grandmont, 37200, Tours, France.
| |
Collapse
|
23
|
Grossert A, Mehrjardi NZ, Bailey SJ, Lindsay MA, Hescheler J, Šarić T, Teusch N. Ketamine Increases Proliferation of Human iPSC-Derived Neuronal Progenitor Cells via Insulin-Like Growth Factor 2 and Independent of the NMDA Receptor. Cells 2019; 8:cells8101139. [PMID: 31554266 PMCID: PMC6830315 DOI: 10.3390/cells8101139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/22/2022] Open
Abstract
The N-methyl-D-aspartate (NMDA) receptor antagonist ketamine offers promising perspectives for the treatment of major depressive disorder. Although ketamine demonstrates rapid and long-lasting effects, even in treatment-resistant patients, to date, the underlying mode of action remains elusive. Thus, the aim of our study was to investigate the molecular mechanism of ketamine at clinically relevant concentrations by establishing an in vitro model based on human induced pluripotent stem cells (iPSCs)-derived neural progenitor cells (NPCs). Notably, ketamine increased the proliferation of NPCs independent of the NMDA receptor, while transcriptome analysis revealed significant upregulation of insulin-like growth factor 2 (IGF2) and p11, a member of the S100 EF-hand protein family, which are both implicated in the pathophysiology of depression, 24 h after ketamine treatment. Ketamine (1 µM) was able to increase cyclic adenosine monophosphate (cAMP) signaling in NPCs within 15 min and cell proliferation, while ketamine-induced IGF2 expression was reduced after PKA inhibition with cAMPS-Rp. Furthermore, 24 h post-administration of ketamine (15 mg/kg) in vivo confirmed phosphorylation of extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) in the subgranular zone (SGZ) of the hippocampus in C57BL/6 mice. In conclusion, ketamine promotes the proliferation of NPCs presumably by involving cAMP-IGF2 signaling.
Collapse
Affiliation(s)
- Alessandra Grossert
- Bio-Pharmaceutical Chemistry and Molecular Pharmacology, Faculty of Applied Natural Sciences, Technische Hochschule Köln, 51373 Leverkusen, Germany.
| | - Narges Zare Mehrjardi
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, 50931 Cologne, Germany.
| | - Sarah J Bailey
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, UK.
| | - Mark A Lindsay
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, UK.
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, 50931 Cologne, Germany.
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, 50931 Cologne, Germany.
| | - Nicole Teusch
- Bio-Pharmaceutical Chemistry and Molecular Pharmacology, Faculty of Applied Natural Sciences, Technische Hochschule Köln, 51373 Leverkusen, Germany.
- Department of Biomedical Sciences, Faculty of Human Sciences, University of Osnabrück, 49076 Osnabrück, Germany.
| |
Collapse
|
24
|
Liu G, Wang Y, Zheng W, Cheng H, Zhou R. P11 Loss-of-Function is Associated with Decreased Cell Proliferation and Neurobehavioral Disorders in Mice. Int J Biol Sci 2019; 15:1383-1395. [PMID: 31337969 PMCID: PMC6643149 DOI: 10.7150/ijbs.33773] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 03/09/2019] [Indexed: 12/20/2022] Open
Abstract
Although depression is associated with anxiety and memory deficit in humans, the molecular mechanisms of the complication remain largely unknown. In this study, we generated P11 knockout mice using CRISPR/Cas9 technology, as well as P11 knockout MEF cell lines, and confirmed depression-like phenotype. We observed that knockout of P11 in MEFs led to a decreased cell proliferation compared with P11+/+ MEFs. Moreover, P11 knockout resulted in a larger cell size, which resulted probably from accumulated F-actin stress fibers. The number of proliferating cells was decreased in the hippocampus of P11 KO mice. We observed anxiety-like disorder in addition to depression phenotype in the knockout mice. In addition, knockout of P11 led to memory deficit in female mice, but not in males. These data indicated that P11 is involved in regulating cell proliferation and cell size. The molecular associations of depression behavior with anxiety and memory deficit suggested a potential approach to improve therapeutic intervention through P11 in these disorders.
Collapse
Affiliation(s)
| | | | | | - Hanhua Cheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Rongjia Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
25
|
Tantyo NA, Karyadi AS, Rasman SZ, Salim MRG, Devina A, Sumarpo A. The prognostic value of S100A10 expression in cancer. Oncol Lett 2018; 17:1417-1424. [PMID: 30675195 PMCID: PMC6341771 DOI: 10.3892/ol.2018.9751] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 11/15/2018] [Indexed: 12/30/2022] Open
Abstract
S100A10, a member of the S100 protein family, commonly forms a heterotetrameric complex with Annexin A2. This is essential for the generation of cellular plasmin from plasminogen, which leads to a cascade of molecular events crucial for tumor progression. S100A10 upregulation has been reported in a number of cancers, suggesting that it may have potential as a prognostic biomarker, as well as predicting sensitivity to anticancer drugs. This review evaluates the direct and indirect relationships between S100A10 and cancer progression by investigating its role in cancer. Research papers published on PubMed and Google Scholar between 2007–2017 were collated and reviewed. We concluded that S100A10 affects the development of the hallmarks of cancer as explained by Hanahan and Weinberg in 2011, most notably by activating the invasion and metastasis of cancer cells. However, further studies are required to explore the underlying biological mechanisms of S100A10.
Collapse
Affiliation(s)
- Normastuti Adhini Tantyo
- Department of Biomedicine, Indonesia International Institute for Life Sciences, Jakarta Timur 13210, Indonesia
| | - Azrina Saraswati Karyadi
- Department of Biomedicine, Indonesia International Institute for Life Sciences, Jakarta Timur 13210, Indonesia
| | - Siti Zulimas Rasman
- Department of Biomedicine, Indonesia International Institute for Life Sciences, Jakarta Timur 13210, Indonesia
| | | | - Astrella Devina
- Department of Biomedicine, Indonesia International Institute for Life Sciences, Jakarta Timur 13210, Indonesia
| | - Anton Sumarpo
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta Utara 14440, Indonesia
| |
Collapse
|
26
|
p11 in Cholinergic Interneurons of the Nucleus Accumbens Is Essential for Dopamine Responses to Rewarding Stimuli. eNeuro 2018; 5:eN-NWR-0332-18. [PMID: 30417079 PMCID: PMC6223111 DOI: 10.1523/eneuro.0332-18.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/04/2018] [Accepted: 10/09/2018] [Indexed: 01/04/2023] Open
Abstract
A recent study showed that p11 expressed in cholinergic interneurons (CINs) of the nucleus accumbens (NAc) is a key regulator of depression-like behaviors. Dopaminergic neurons projecting to the NAc are responsible for reward-related behaviors, and their function is impaired in depression. The present study investigated the role of p11 in NAc CINs in dopamine responses to rewarding stimuli. The extracellular dopamine and acetylcholine (ACh) levels in the NAc were determined in freely moving male mice using in vivo microdialysis. Rewarding stimuli (cocaine, palatable food, and female mouse encounter) induced an increase in dopamine efflux in the NAc of wild-type (WT) mice. The dopamine responses were attenuated (cocaine) or abolished (food and female mouse encounter) in constitutive p11 knock-out (KO) mice. The dopamine response to cocaine was accompanied by an increase in ACh NAc efflux, whereas the attenuated dopamine response to cocaine in p11 KO mice was restored by activation of nicotinic or muscarinic ACh receptors in the NAc. Dopamine responses to rewarding stimuli and ACh release in the NAc were attenuated in mice with deletion of p11 from cholinergic neurons (ChAT-p11 cKO mice), whereas gene delivery of p11 to CINs restored the dopamine responses. Furthermore, chemogenetic studies revealed that p11 is required for activation of CINs in response to rewarding stimuli. Thus, p11 in NAc CINs plays a critical role in activating these neurons to mediate dopamine responses to rewarding stimuli. The dysregulation of mesolimbic dopamine system by dysfunction of p11 in NAc CINs may be involved in pathogenesis of depressive states.
Collapse
|
27
|
Gestational stress in mouse dams negatively affects gestation and postpartum hippocampal BDNF and P11 protein levels. Mol Cell Neurosci 2018; 88:292-299. [DOI: 10.1016/j.mcn.2018.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/31/2018] [Accepted: 02/26/2018] [Indexed: 11/23/2022] Open
|
28
|
Liang J, Yue Y, Jiang H, Geng D, Wang J, Lu J, Li S, Zhang K, Wu A, Yuan Y. Genetic variations in the p11/tPA/BDNF pathway are associated with post stroke depression. J Affect Disord 2018; 226:313-325. [PMID: 29028593 DOI: 10.1016/j.jad.2017.09.055] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 08/20/2017] [Accepted: 09/27/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND The effects of BDNF on post stroke depression (PSD) may be influenced by genetic variations in intracellular signal transduction pathways, such as the p11/tPA/BDNF pathway. In this study, we aimed to determine the association of polymorphisms in candidate genes of the gene transduction pathway with PSD, as well as the effects of the interactions between genes in our Chinese sample. METHODS Two-hundred-fifty-four Chinese samples with acute ischaemic stroke included 122 PSD patients and 132 nonPSD patients. Sixty-five single nucleotide polymorphisms (SNPs) in six genes (p11, tPA, PAI-1, BDNF, TrkB and p75NTR) of the p11/tPA/BDNF pathway with minor allele frequencies > 5% were successfully genotyped from an initial series of 76 SNPs. The severity of depressive symptoms was assessed by the 17-item Hamilton Depression Rating scale score. Environmental factors were measured with the life events scale and social support rating scale for all patients. SNP and haplotype associations were analysed using gPLINK software. Gene-gene interactions were evaluated with generalized multifactor dimensionality reduction software. RESULTS The results showed that TrkB polymorphisms (rs11140793AC genotype, rs7047042CG genotype, rs1221CT genotype, rs2277193TC genotype and rs2277192AG genotype) were significantly associated with PSD. Three haplotypes (AT, GG, and AAT) of TrkB were significantly associated with PSD. Seven haplotypes (GC, AG, ACG, CGC, GCT, ACGC and ACAT) of BDNF were significantly correlated with PSD. We identified significant gene-gene interactions between the p11 (rs11204922 SNP), tPA (rs8178895, rs2020918 SNPs) and BDNF (rs6265, rs2049046, rs16917271, rs727155 SNPs) genes in the PSD group. We also identified significant gene-gene interactions between the BDNF (rs2049046, rs7931247 SNPs) and TrkB (rs7816 SNP) genes with increased occurrence of PSD and sig gene-gene interactions between the BDNF gene (rs6265, rs56164415, rs2049046, rs4923468, rs2883187, rs16917271, rs1491850, rs727155, rs2049048 SNPs) and p75NTR gene (rs2072446, rs11466155) in the PSD group. CONCLUSION These findings provides evidence that the TrkB gene, BDNF and TrkB haplotypes, and gene-gene interactions between p11, tPA and BDNF are all associated with PSD, which suggests that genetic variations in the p11/tPA/BDNF pathway may play a central role in regulating the underlying mechanism of PSD.
Collapse
Affiliation(s)
- Jinfeng Liang
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, Medical School of Southeast University, Nanjing 210009, PR China; Institute of Psychosomatics, Medical School of Southeast University, Nanjing 210009, PR China
| | - Yingying Yue
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, Medical School of Southeast University, Nanjing 210009, PR China; Institute of Psychosomatics, Medical School of Southeast University, Nanjing 210009, PR China
| | - Haitang Jiang
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, Medical School of Southeast University, Nanjing 210009, PR China; Institute of Psychosomatics, Medical School of Southeast University, Nanjing 210009, PR China
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221000, PR China
| | - Jun Wang
- Department of Neurology, Nanjing First Hospital, Nanjing 210006, PR China
| | - Jianxin Lu
- Department of Neurology, The Peoples' Hospital of Gaochun County, Nanjing 211300, PR China
| | - Shenghua Li
- Department of Neurology, Jiangning Nanjing hospital, Nanjing 211100, PR China
| | - Kezhong Zhang
- Department of Neurology, the Affiliated First hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Aiqin Wu
- Department of Psychosomatics, the First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, Medical School of Southeast University, Nanjing 210009, PR China; Institute of Psychosomatics, Medical School of Southeast University, Nanjing 210009, PR China.
| |
Collapse
|
29
|
Mooney SJ, Nobrega JN, Levitt AJ, Hynynen K. Antidepressant effects of focused ultrasound induced blood-brain-barrier opening. Behav Brain Res 2018; 342:57-61. [PMID: 29326057 DOI: 10.1016/j.bbr.2018.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/05/2018] [Accepted: 01/06/2018] [Indexed: 01/18/2023]
Abstract
In many cases, hippocampal neurogenesis appears to be a hallmark of antidepressant treatments. One novel technique for inducing this type of neurogenesis is using focused ultrasound waves, in conjunction with circulating microbubbles, to open the blood-brain-barrier. The present experiment aimed to test whether this technique has antidepressant effects in a rodent model. Rats were subjected to 1, 2 or 3 weekly treatments of magnetic resonance-guided focused ultrasound in order to open the blood-brain-barrier in the hippocampal region. Before and after treatments, animals went through modified forced swim tests. 1 week after the final treatment, animals that received 2 weekly treatments showed antidepressant-like effects on behavioural measures in comparison to untreated controls. This was not the case for animals that received 1 or 3 weekly treatments. Effects had disappeared by 5 weeks following the first ultrasound treatment. These results suggest that focused ultrasound may be used for inducing short-term antidepressant effects.
Collapse
Affiliation(s)
- Skyler J Mooney
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada.
| | - José N Nobrega
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Behavioural Neurobiology Laboratory, Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Anthony J Levitt
- Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
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
|
31
|
Mitic M, Brkic Z, Lukic I, Adzic M. Convergence of glycogen synthase kinase 3β and GR signaling in response to fluoxetine treatment in chronically stressed female and male rats. Behav Brain Res 2017; 333:295-303. [PMID: 28729116 DOI: 10.1016/j.bbr.2017.07.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/07/2017] [Accepted: 07/14/2017] [Indexed: 10/19/2022]
Abstract
Accumulating evidence strongly suggest that impaired glucocorticoid receptor (GR) signaling is involved in stress-related mood disorders, and nominate GR as a potential target for antidepressants (ADs). It is known that different classes of ADs affects the GR action via modifying its phosphorylation, while the mechanism through which ADs alter GR phosphorylation targeted by GSK3β, a kinase modulated via serotonin neurotransmission, are unclear. On this basis, we investigated whether GSK3β-GR signaling could be a convergence point of fluoxetine action on brain function and behavior, by examining its effect on GSK3β targeted-GR phosphorylation on threonine 171 (pGR171), and expression of GR-regulated genes in the hippocampus of female and male rats exposed to chronic isolation stress. Stress induced sex-specific GSK3β-targeted phosphorylation of pGR171 in the nucleus of the hippocampus of stressed animals. Namely, while in females stress triggered coupled action of GSK3β-pGR171 signaling, in males changes in pGR171 levels did not correspond to GSK3β activity. On the other hand, fluoxetine managed to up-regulate this pathway in sex-unbiased manner. Furthermore, fluoxetine reverted stress-induced changes in most of the analyzed genes in males, CRH, 5-HT1a and p11, while in females its effect was limited to CRH. These data further suggest that pGR171 signaling affects cellular localization of GR in response to chronic stress and fluoxetine in both sexes. Collectively, our results describe a novel convergence point between GR signaling and GSK3β pathway in rat hippocampus in response to stress and fluoxetine in both sexes and its involvement in fluoxetine-regulated brain function in males.
Collapse
Affiliation(s)
- Milos Mitic
- Department of Molecular Biology and Endocrinology, University of Belgrade, Vinca Institute of Nuclear Sciences, P.O. Box-522-MBE090, 11001 Belgrade, Serbia.
| | - Zeljka Brkic
- Department of Molecular Biology and Endocrinology, University of Belgrade, Vinca Institute of Nuclear Sciences, P.O. Box-522-MBE090, 11001 Belgrade, Serbia
| | - Iva Lukic
- Department of Molecular Biology and Endocrinology, University of Belgrade, Vinca Institute of Nuclear Sciences, P.O. Box-522-MBE090, 11001 Belgrade, Serbia; Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Miroslav Adzic
- Department of Molecular Biology and Endocrinology, University of Belgrade, Vinca Institute of Nuclear Sciences, P.O. Box-522-MBE090, 11001 Belgrade, Serbia
| |
Collapse
|
32
|
Medrihan L, Sagi Y, Inde Z, Krupa O, Daniels C, Peyrache A, Greengard P. Initiation of Behavioral Response to Antidepressants by Cholecystokinin Neurons of the Dentate Gyrus. Neuron 2017; 95:564-576.e4. [DOI: 10.1016/j.neuron.2017.06.044] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 03/09/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022]
|
33
|
King JR, Velasquez JC, Torii M, Bonnin A. Effect of Maternal ±Citalopram Exposure on P11 Expression and Neurogenesis in the Mouse Fetal Brain. ACS Chem Neurosci 2017; 8:1019-1025. [PMID: 28076682 DOI: 10.1021/acschemneuro.6b00339] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Fetal exposure to selective serotonin reuptake inhibitors (SSRI) has been associated with increased risk of adverse neurodevelopmental outcomes. In the adult brain, SSRI therapy regulates p11 (s100a10) expression and alters neurogenesis. The protein p11 indirectly regulates 5-HT signaling through 5-HT1B/D receptors. In the fetal brain, signaling through these receptors modulates axonal circuit formation. We determined whether p11 is expressed in the fetal mouse brain, and whether maternal SSRI exposure affects fetal p11 expression and neurogenesis. The SSRI ± citalopram was administered to pregnant mice from gestational day 8 to 17. Results show that p11 is expressed in fetal thalamic neurons and thalamocortical axons. Furthermore, p11 protein expression is significantly decreased in the fetal thalamus after in utero ±citalopram exposure compared to untreated controls, and neurogenesis is significantly decreased in specific fetal brain regions. These findings reveal differential regulation of p11 expression and altered neurogenesis in the fetal brain as a result of maternal SSRI exposure.
Collapse
Affiliation(s)
- Jennifer R. King
- Department
of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine,
Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
| | - Juan C. Velasquez
- Zilkha
Neurogenetic Institute and Department of Cell and Neurobiology, Keck School of Medicine of University of Southern California, Los Angeles, California 90089, United States
| | - Masaaki Torii
- Center
for Neuroscience Research, Children’s National Medical Center, Washington,
D.C. 20010, United States
- Department
of Pediatrics, Pharmacology and Physiology, The George Washington University, Washington, D.C. 20052, United States
| | - Alexandre Bonnin
- Zilkha
Neurogenetic Institute and Department of Cell and Neurobiology, Keck School of Medicine of University of Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
34
|
Egeland M, Guinaudie C, Du Preez A, Musaelyan K, Zunszain PA, Fernandes C, Pariante CM, Thuret S. Depletion of adult neurogenesis using the chemotherapy drug temozolomide in mice induces behavioural and biological changes relevant to depression. Transl Psychiatry 2017; 7:e1101. [PMID: 28440814 PMCID: PMC5416706 DOI: 10.1038/tp.2017.68] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 02/15/2017] [Accepted: 02/18/2017] [Indexed: 12/17/2022] Open
Abstract
Numerous studies have examined links between postnatal neurogenesis and depression using a range of experimental methods to deplete neurogenesis. The antimitotic drug temozolomide (TMZ) has previously been used successfully as an experimental tool in animals to deplete adult neurogenesis and is used regularly on human patients as a standard chemotherapy for brain cancer. In this study, we wanted to evaluate whether TMZ as a model for chemotherapy treatment could affect parameters related to depression in an animal model. Prevalence rates of depression in patients is thought to be highly underdiagnosed, with some studies reporting rates as high as 90%. Results from this study in mice, treated with a regimen of TMZ similar to humans, exhibited behavioural and biochemical changes that have relevance to the development of depression. In particular, behavioural results demonstrated robust deficits in processing novelty and a significant increase in the corticosterone response. Quantification of neurogenesis using a novel sectioning method, which clearly evaluates dorsal and ventral neurogenesis separately, showed a significant correlation between the level of ventral neurogenesis and the corticosterone response. Depression is a complex disorder with discoveries regarding its neurobiology and how it relates to behaviour being only in their infancy. The findings presented in this study demonstrate that chemotherapy-induced decreases in neurogenesis results in previously unreported behavioural and biochemical consequences. These results, we argue, are indicative of a biological mechanism, which may contribute to the development of depression in patients being treated with chemotherapy and is separate from the mental distress resulting from a cancer diagnosis.
Collapse
Affiliation(s)
- M Egeland
- Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK,Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK,Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Cutcombe Road, London SE5 9RT, UK. E-mail: or
| | - C Guinaudie
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - A Du Preez
- Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK,MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - K Musaelyan
- Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK,Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK,MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - P A Zunszain
- Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - C Fernandes
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - C M Pariante
- Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - S Thuret
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK,Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Cutcombe Road, London SE5 9RT, UK. E-mail: or
| |
Collapse
|
35
|
Milosevic A, Liebmann T, Knudsen M, Schintu N, Svenningsson P, Greengard P. Cell- and region-specific expression of depression-related protein p11 (S100a10) in the brain. J Comp Neurol 2016; 525:955-975. [PMID: 27616678 PMCID: PMC5222728 DOI: 10.1002/cne.24113] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 08/03/2016] [Accepted: 08/09/2016] [Indexed: 12/20/2022]
Abstract
P11 (S100a10), a member of the S100 family of proteins, has widespread distribution in the vertebrate body, including in the brain, where it has a key role in membrane trafficking, vesicle secretion, and endocytosis. Recently, our laboratory has shown that a constitutive knockout of p11 (p11-KO) in mice results in a depressive-like phenotype. Furthermore, p11 has been implicated in major depressive disorder (MDD) and in the actions of antidepressants. Since depression affects multiple brain regions, and the role of p11 has only been determined in a few of these areas, a detailed analysis of p11 expression in the brain is warranted. Here we demonstrate that, although widespread in the brain, p11 expression is restricted to distinct regions, and specific neuronal and nonneuronal cell types. Furthermore, we provide comprehensive mapping of p11 expression using in situ hybridization, immunocytochemistry, and whole-tissue volume imaging. Overall, expression spans multiple brain regions, structures, and cell types, suggesting a complex role of p11 in depression. J. Comp. Neurol. 525:955-975, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ana Milosevic
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York, USA
| | - Thomas Liebmann
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York, USA
| | - Margarete Knudsen
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York, USA
| | - Nicoletta Schintu
- Section for Translational Neuropharmacology, Department of Clinical Neuroscience, CMM L8:01, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Per Svenningsson
- Section for Translational Neuropharmacology, Department of Clinical Neuroscience, CMM L8:01, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York, USA
| |
Collapse
|
36
|
Park SW, Nhu LH, Cho HY, Seo MK, Lee CH, Ly NN, Choi CM, Lee BJ, Kim GM, Seol W, Lee JG, Kim YH. p11 mediates the BDNF-protective effects in dendritic outgrowth and spine formation in B27-deprived primary hippocampal cells. J Affect Disord 2016; 196:1-10. [PMID: 26896741 DOI: 10.1016/j.jad.2016.02.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 10/28/2015] [Accepted: 02/07/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND p11 (S100A10) is a key regulator of depression-like behaviors and antidepressant drug response in rodent models. Recent studies suggest that p11 mediates the behavioral antidepressant action of brain-derived neurotrophic factor (BDNF) in rodents. BDNF improves neural plasticity, which is linked to the cellular actions of antidepressant drugs. In the present study, we investigated whether p11 regulated BDNF action on neural plasticity in vitro. METHODS We generated primary hippocampal cultures. p11 expression, total dendritic length, and spine density were investigated under toxic conditions induced by B27 deprivation, which causes hippocampal cell death. RESULTS B27 deprivation significantly decreased p11 expression. Treatment with BDNF significantly prevented the B27 deprivation-induced decrease in p11 levels in a concentration-dependent manner, whereas these concentrations had no effect on control cultures. B27 deprivation significantly reduced the total length of hippocampal dendrites and spine density. BDNF increased the total dendritic length and spine density in conditions with or without B27. Furthermore, p11 knockdown through small interfering RNA (siRNA) transfection blocked these effects. The overexpression of p11 in B27-deprived cells increased the total dendritic length and spine density, and treatment with BDNF potentiated these effects. LIMITATION This study should be confirmed in animal models of depression. CONCLUSION Taken together, our data suggest that BDNF-induced improvement in neural plasticity may depend on the regulation of p11 in hippocampal cells with B27 deprivation. These results provide evidence to strengthen the theoretical basis of a role for p11 in BDNF-induced antidepressant action.
Collapse
Affiliation(s)
- Sung Woo Park
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea; Department of health science and technology, Graduate School of Inje University, Busan, Republic of Korea
| | - Le Hoa Nhu
- Department of health science and technology, Graduate School of Inje University, Busan, Republic of Korea
| | - Hye Yeon Cho
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| | - Mi Kyoung Seo
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| | - Chan Hong Lee
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| | - Nguyen Ngoc Ly
- Department of health science and technology, Graduate School of Inje University, Busan, Republic of Korea
| | - Cheol Min Choi
- Department of health science and technology, Graduate School of Inje University, Busan, Republic of Korea
| | - Bong Ju Lee
- Department of Psychiatry, School of Medicine, Haeundae Paik Hospital, Inje University, Busan, Republic of Korea
| | - Gyung-Mee Kim
- Department of Psychiatry, School of Medicine, Haeundae Paik Hospital, Inje University, Busan, Republic of Korea
| | - Wongi Seol
- InAm Neuroscience Research Center, Wonkwang University, Sanbon Hospital, Gunpo, kyeonggi-do, Republic of Korea
| | - Jung Goo Lee
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea; Department of health science and technology, Graduate School of Inje University, Busan, Republic of Korea; Department of Psychiatry, School of Medicine, Haeundae Paik Hospital, Inje University, Busan, Republic of Korea.
| | - Young Hoon Kim
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea; Department of health science and technology, Graduate School of Inje University, Busan, Republic of Korea; Department of Psychiatry, School of Medicine, Haeundae Paik Hospital, Inje University, Busan, Republic of Korea.
| |
Collapse
|
37
|
Enzymatic Depletion of the Polysialic Acid Moiety Associated with the Neural Cell Adhesion Molecule Inhibits Antidepressant Efficacy. Neuropsychopharmacology 2016; 41:1670-80. [PMID: 26530284 PMCID: PMC4832030 DOI: 10.1038/npp.2015.337] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/04/2015] [Accepted: 10/27/2015] [Indexed: 01/01/2023]
Abstract
Antidepressant drugs are too often ineffective, the exact mechanism of efficacy is still ambiguous, and there has been a paucity of novel targets for pharmacotherapy. In an attempt to understand the pathogenesis of depression and subsequently develop more efficacious antidepressant drugs, multiple theories have been proposed, including the modulation of neurotransmission, the upregulation of neurogenesis and neurotrophic factors, normalizing hypothalamic-pituitary-adrenal reactivity, and the reduction of neuroinflammation; all of which have supporting lines of evidence. Therefore, an ideal molecular target for novel pharmaceutical intervention would function at the confluence of these theories. The polysialylated form of the neural cell adhesion molecule (PSA-NCAM) functions broadly, serving to mediate synaptic plasticity, neurogenesis, neurotrophic factor signaling, and inflammatory signaling throughout the brain; all of which are associated with the pathophysiology and treatment of depression. Moreover, the expression of PSA-NCAM is reduced by depression, and conversely enhanced by antidepressant treatment, particularly within the hippocampus. Here we demonstrate that selectively cleaving the polysialic acid moiety, using the bacteriophage-derived enzyme endoneuraminidase N, completely inhibits the antidepressant efficacy of the selective-serotonin reuptake inhibitor fluoxetine (FLX) in a chronic unpredictable stress model of depression. We also observe a corresponding attenuation of FLX-induced hippocampal neuroplasticity, including decreased hippocampal neurogenesis, synaptic density, and neural activation. These data indicate that PSA-NCAM-mediated neuroplasticity is necessary for antidepressant action; therefore PSA-NCAM represents an interesting, and novel, target for pharmacotherapy.
Collapse
|
38
|
Theilmann W, Kleimann A, Rhein M, Bleich S, Frieling H, Löscher W, Brandt C. Behavioral differences of male Wistar rats from different vendors in vulnerability and resilience to chronic mild stress are reflected in epigenetic regulation and expression of p11. Brain Res 2016; 1642:505-515. [PMID: 27103570 DOI: 10.1016/j.brainres.2016.04.041] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 04/15/2016] [Accepted: 04/16/2016] [Indexed: 11/27/2022]
Abstract
Outbred rat lines such as Wistar rats are commonly used for models of depressive disorders. Such rats arise from random mating schedules. Hence, genetic drift occurs in outbred populations which could lead to genotypic and phenotypic heterogeneity between rats from different vendors. Additionally, vendor specific rearing conditions could contribute to intrastrain variability. In the present study differences in behavioral responses to the chronic mild stress (CMS) model of depression within Wistar rat strains from different vendors are described. DNA methylation studies and mRNA expression analysis of p11 revealed that the behavioral differences between the substrains are reflected at the epigenetic and genetic level. The results suggest that there are breeder-dependent differences in vulnerability to stress in the CMS model of depression, which might bear on the validity of the model and contribute to contradictory findings and difficulties of replication between laboratories. P11 mRNA expression seems to be differently regulated depending on the quality of the stress response evoked by CMS exposure.
Collapse
Affiliation(s)
- Wiebke Theilmann
- Neuroscience Center, University of Helsinki, Finland; Center for Systems Neuroscience, Hannover, Germany
| | - Alexandra Kleimann
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover, Germany
| | - Mathias Rhein
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover, Germany
| | - Stefan Bleich
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Helge Frieling
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Medical School Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Claudia Brandt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
39
|
Sun HL, Zhou ZQ, Zhang GF, Yang C, Wang XM, Shen JC, Hashimoto K, Yang JJ. Role of hippocampal p11 in the sustained antidepressant effect of ketamine in the chronic unpredictable mild stress model. Transl Psychiatry 2016; 6:e741. [PMID: 26905413 PMCID: PMC4872431 DOI: 10.1038/tp.2016.21] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 12/15/2015] [Accepted: 01/26/2016] [Indexed: 02/07/2023] Open
Abstract
Although ketamine shows a rapid and sustained antidepressant effect, the precise mechanisms underlying its effect are unknown. Recent studies indicate a key role of p11 (also known as S100A10) in depression-like behavior in rodents. The present study aimed to investigate the role of p11 in the antidepressant-like action of ketamine in chronic unpredictable mild stress (CUMS) rat model. The open-field test, forced swimming test and sucrose preference test were performed after administration of ketamine (10 mg kg(-1)) or a combination of ketamine and ANA-12 (a tropomyosin-related kinase B (TrkB) antagonist; 0.5 mg kg(-1)). The lentivirus vector for p11 was constructed to knock down the hippocampal expression of p11. In the CUMS rats, ketamine showed a rapid (0.5 h) and sustained (72 h) antidepressant effect, and its effect was significantly blocked by co-administration of ANA-12. Furthermore, ketamine significantly increased the reduced expression of brain-derived neurotrophic factor (BDNF) in the hippocampus of CUMS rats, whereas ketamine did not affect the expression of p11 in CUMS rats 0.5 h after administration. In addition, ketamine significantly increased the reduced ratio of p-TrkB/TrkB in the hippocampus by CUMS rats, and its effect was also blocked by ANA-12. Moreover, the reduced expression of BDNF and p11 in the hippocampus of CUMS rats was significantly recovered to control levels 72 h after ketamine administration. Interestingly, knockdown of hippocampal p11 caused increased immobility time and decreased sucrose preference, which were not improved by ketamine administration. These results suggest that p11 in the hippocampus may have a key role in the sustained antidepressant effect of ketamine in the CUMS model of depression.
Collapse
Affiliation(s)
- H-L Sun
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Z-Q Zhou
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - G-F Zhang
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - C Yang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - X-M Wang
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - J-C Shen
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - K Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan,Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, 1-8-1 Inohana, Chiba 260-8670, Japan E-mail:
| | - J-J Yang
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China,Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, China. E-mail:
| |
Collapse
|
40
|
Probable involvement of p11 with interferon alpha induced depression. Sci Rep 2016; 6:17029. [PMID: 26821757 PMCID: PMC4731785 DOI: 10.1038/srep17029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 10/23/2015] [Indexed: 12/16/2022] Open
Abstract
Depression is one of the major side effects of interferon alpha (IFN-α) treatment, but the molecular mechanism underlying IFN-α-induced depression remains unclear. Several studies have shown that the serotonin receptors 5-HTR1b and 5-HTR4 play key roles in the anti-depression effects associated with p11 (S100A10). We investigated the effects of IFN-α on the regulation of p11, 5-HTR1b and 5-HTR4 in mice and human neuroblastoma cells (SH-sy5y). We found that intraperitoneal injection with IFN-α in Balb/c mice resulted in an increased immobility in FST and TST, and potently lowered the protein levels of p11, 5-HTR1b and 5-HTR4 in the hippocampus or cingulate gyrus. IFN-α significantly down-regulated the protein levels of p11, 5-HTR1b and 5-HTR4 in SH-sy5y cells, in a time- and dose-dependent manner. Our study revealed that over-expression of p11 could prevent the IFN-α-induced down-regulation of 5-HTR1b and 5-HTR4. The results indicated that IFN-α treatment resulted in p11 down-regulation, which subsequently decreased 5-HTR1b and 5-HTR4 in vitro or in vivo. Our findings suggested that p11 might be a potential regulator on 5-HTR1b and 5-HTR4 as well as a predictor of or a therapeutic target for IFN-α-induced depression.
Collapse
|
41
|
Alteration by p11 of mGluR5 localization regulates depression-like behaviors. Mol Psychiatry 2015; 20:1546-56. [PMID: 26370144 PMCID: PMC4907335 DOI: 10.1038/mp.2015.132] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 05/26/2015] [Accepted: 06/16/2015] [Indexed: 12/19/2022]
Abstract
Mood disorders and antidepressant therapy involve alterations of monoaminergic and glutamatergic transmission. The protein S100A10 (p11) was identified as a regulator of serotonin receptors, and it has been implicated in the etiology of depression and in mediating the antidepressant actions of selective serotonin reuptake inhibitors. Here we report that p11 can also regulate depression-like behaviors via regulation of a glutamatergic receptor in mice. p11 directly binds to the cytoplasmic tail of metabotropic glutamate receptor 5 (mGluR5). p11 and mGluR5 mutually facilitate their accumulation at the plasma membrane, and p11 increases cell surface availability of the receptor. Whereas p11 overexpression potentiates mGluR5 agonist-induced calcium responses, overexpression of mGluR5 mutant, which does not interact with p11, diminishes the calcium responses in cultured cells. Knockout of mGluR5 or p11 specifically in glutamatergic neurons in mice causes depression-like behaviors. Conversely, knockout of mGluR5 or p11 in GABAergic neurons causes antidepressant-like behaviors. Inhibition of mGluR5 with an antagonist, 2-methyl-6-(phenylethynyl)pyridine (MPEP), induces antidepressant-like behaviors in a p11-dependent manner. Notably, the antidepressant-like action of MPEP is mediated by parvalbumin-positive GABAergic interneurons, resulting in a decrease of inhibitory neuronal firing with a resultant increase of excitatory neuronal firing. These results identify a molecular and cellular basis by which mGluR5 antagonism achieves its antidepressant-like activity.
Collapse
|
42
|
Scharfman HE, Bernstein HL. Potential implications of a monosynaptic pathway from mossy cells to adult-born granule cells of the dentate gyrus. Front Syst Neurosci 2015; 9:112. [PMID: 26347618 PMCID: PMC4541026 DOI: 10.3389/fnsys.2015.00112] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 07/20/2015] [Indexed: 11/13/2022] Open
Abstract
The dentate gyrus (DG) is important to many aspects of hippocampal function, but there are many aspects of the DG that are incompletely understood. One example is the role of mossy cells (MCs), a major DG cell type that is glutamatergic and innervates the primary output cells of the DG, the granule cells (GCs). MCs innervate the GCs as well as local circuit neurons that make GABAergic synapses on GCs, so the net effect of MCs on GCs – and therefore the output of the DG – is unclear. Here we first review fundamental information about MCs and the current hypotheses for their role in the normal DG and in diseases that involve the DG. Then we review previously published data which suggest that MCs are a source of input to a subset of GCs that are born in adulthood (adult-born GCs). In addition, we discuss the evidence that adult-born GCs may support the normal inhibitory ‘gate’ functions of the DG, where the GCs are a filter or gate for information from the entorhinal cortical input to area CA3. The implications are then discussed in the context of seizures and temporal lobe epilepsy (TLE). In TLE, it has been suggested that the DG inhibitory gate is weak or broken and MC loss leads to insufficient activation of inhibitory neurons, causing hyperexcitability. That idea was called the “dormant basket cell hypothesis.” Recent data suggest that loss of normal adult-born GCs may also cause disinhibition, and seizure susceptibility. Therefore, we propose a reconsideration of the dormant basket cell hypothesis with an intervening adult-born GC between the MC and basket cell and call this hypothesis the “dormant immature granule cell hypothesis.”
Collapse
Affiliation(s)
- Helen E Scharfman
- The Nathan Kline Institute for Psychiatric Research, Orangeburg NY, USA ; New York University Langone Medical Center, New York NY, USA
| | - Hannah L Bernstein
- The Nathan Kline Institute for Psychiatric Research, Orangeburg NY, USA ; New York University Langone Medical Center, New York NY, USA
| |
Collapse
|
43
|
Caldarone BJ, Zachariou V, King SL. Rodent models of treatment-resistant depression. Eur J Pharmacol 2014; 753:51-65. [PMID: 25460020 DOI: 10.1016/j.ejphar.2014.10.063] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 09/16/2014] [Accepted: 10/09/2014] [Indexed: 01/06/2023]
Abstract
Major depression is a prevalent and debilitating disorder and a substantial proportion of patients fail to reach remission following standard antidepressant pharmacological treatment. Limited efficacy with currently available antidepressant drugs highlights the need to develop more effective medications for treatment- resistant patients and emphasizes the importance of developing better preclinical models that focus on treatment- resistant populations. This review discusses methods to adapt and refine rodent behavioral models that are predictive of antidepressant efficacy to identify populations that show reduced responsiveness or are resistant to traditional antidepressants. Methods include separating antidepressant responders from non-responders, administering treatments that render animals resistant to traditional pharmacological treatments, and identifying genetic models that show antidepressant resistance. This review also examines pharmacological and non-pharmacological treatments regimes that have been effective in refractory patients and how some of these approaches have been used to validate animal models of treatment-resistant depression. The goals in developing rodent models of treatment-resistant depression are to understand the neurobiological mechanisms involved in antidepressant resistance and to develop valid models to test novel therapies that would be effective in patients that do not respond to traditional monoaminergic antidepressants.
Collapse
Affiliation(s)
- Barbara J Caldarone
- Department of Neurology, Brigham and Women's Hospital and NeuroBehavior Laboratory, Harvard NeuroDiscovery Center, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| | - Venetia Zachariou
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY 10029, USA
| | - Sarah L King
- School of Psychology, University of Sussex, Brighton, East Sussex, UK
| |
Collapse
|
44
|
Svenningsson P, Berg L, Matthews D, Ionescu DF, Richards E, Niciu M, Malinger A, Toups M, Manji H, Trivedi MH, Zarate CA, Greengard P. Preliminary evidence that early reduction in p11 levels in natural killer cells and monocytes predicts the likelihood of antidepressant response to chronic citalopram. Mol Psychiatry 2014; 19:962-4. [PMID: 24614495 PMCID: PMC4161667 DOI: 10.1038/mp.2014.13] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The prediction of treatment response in many neuropsychiatric disorders would be facilitated by easily accessible biomarkers. Using flow cytometry, we herein demonstrate correlations between early reductions of p11 levels in Natural Killer (NK) cells and monocytes and antidepressant response to citalopram in patients with major depressive disorder (MDD).
Collapse
Affiliation(s)
- P Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - L Berg
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - D Matthews
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, Bethesda, USA
| | - DF Ionescu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, Bethesda, USA
| | - E Richards
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, Bethesda, USA
| | - M Niciu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, Bethesda, USA
| | - A Malinger
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, Bethesda, USA
| | - M Toups
- Department of Psychiatry and the Mood Disorders Research Program and Clinic, University of Texas Southwestern Medical Center, Dallas, USA
| | - H Manji
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, Bethesda, USA
| | - MH Trivedi
- Department of Psychiatry and the Mood Disorders Research Program and Clinic, University of Texas Southwestern Medical Center, Dallas, USA
| | - CA Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, Bethesda, USA
| | - P Greengard
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, USA
| |
Collapse
|
45
|
Kong H, Zeng XN, Fan Y, Yuan ST, Ge S, Xie WP, Wang H, Hu G. Aquaporin-4 knockout exacerbates corticosterone-induced depression by inhibiting astrocyte function and hippocampal neurogenesis. CNS Neurosci Ther 2014; 20:391-402. [PMID: 24422972 DOI: 10.1111/cns.12222] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 12/03/2013] [Accepted: 12/03/2013] [Indexed: 02/06/2023] Open
Abstract
AIMS The predominant expression of aquaporin-4 (AQP4) in the brain implies that this water channel may be involved in a range of brain disorders. This study was designed to investigate the role of AQP4 in the pathogenesis of depression, and related possible biological mechanism. METHODS AND RESULTS Wild-type (AQP4(+/+) ) and AQP4 knockout (AQP4(-/-) ) mice were given daily subcutaneous injections of corticosterone (20 mg/kg) for consecutive 21 days. Forced swimming test (FST) and tail suspension test (TST) showed longer immobility times in corticosterone-treated AQP4(-/-) genotype, indicating AQP4 knockout exacerbated depressive-like behaviors in mice. Using immunohistological staining, western blot, and enzyme-linked immunosorbent assay (ELISA), we found a significant loss of astrocytes, aggravated downregulation of excitatory amino acid transporter 2 (EAAT2), synapsin-1, and glial cell line-derived neurotrophic factor (GDNF) in the hippocampus of AQP4(-/-) mice. Moreover, even less hippocampal neurogenesis was identified in corticosterone-treated AQP4(-/-) mice in vivo and hippocampus-derived adult neural stem cells (ANSCs) in vitro. CONCLUSIONS The present findings suggest AQP4 involves the pathogenesis of depression by modulating astrocytic function and adult neurogenesis, highlighting a novel profile of AQP4 as a potential target for the treatment for depression.
Collapse
Affiliation(s)
- Hui Kong
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Adult hippocampal neurogenesis in depression: behavioral implications and regulation by the stress system. Curr Top Behav Neurosci 2014; 18:25-43. [PMID: 24478038 DOI: 10.1007/7854_2014_275] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adult hippocampal neurogenesis, the birth of new neurons in the dentate gyrus of the adult brain, can be regulated by stress and antidepressant treatment, and has consistently been implicated in the behavioral neurobiology of stress-related disorders, especially depression and anxiety. A reciprocal relationship between hippocampal neurogenesis and the hypothalamus-pituitary-adrenal (HPA) axis has recently been suggested, which may play a crucial role in the development and in the resolution of depressive symptoms. This chapter will review some of the existing evidence for stress- and antidepressant-induced changes in adult hippocampal neurogenesis, and critically evaluate the behavioral effects of these changes for depression and anxiety. The potential role of neurogenesis as a neurobiological mechanism for sustained remission from depressive symptoms will be discussed, integrating existing data from clinical studies, animal work, and cellular models. The effect of glucocorticoid hormones and the glucocorticoid receptor (GR) will thereby be evaluated as a central mechanism by which stress and antidepressant may exert their opposing effects on neurogenesis, and ultimately, on mood and behavior.
Collapse
|
47
|
p11 and its role in depression and therapeutic responses to antidepressants. Nat Rev Neurosci 2013; 14:673-80. [PMID: 24002251 DOI: 10.1038/nrn3564] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Studies of the multifunctional protein p11 (also known as S100A10) are shedding light on the molecular and cellular mechanisms underlying depression. Here, we review data implicating p11 in both the amplification of serotonergic signalling and the regulation of gene transcription. We summarize studies demonstrating that levels of p11 are regulated in depression and by antidepressant regimens and, conversely, that p11 regulates depression-like behaviours and/or responses to antidepressants. Current and future studies of p11 may provide a molecular and cellular framework for the development of novel antidepressant therapies.
Collapse
|
48
|
Bidirectional regulation of emotional memory by 5-HT1B receptors involves hippocampal p11. Mol Psychiatry 2013; 18:1096-105. [PMID: 23032875 PMCID: PMC3781317 DOI: 10.1038/mp.2012.130] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 07/16/2012] [Accepted: 07/23/2012] [Indexed: 12/25/2022]
Abstract
Cognitive impairments are common in depression and involve dysfunctional serotonin neurotransmission. The 5-HT1B receptor (5-HT(1B)R) regulates serotonin transmission, via presynaptic receptors, but can also affect transmitter release at heterosynaptic sites. This study aimed at investigating the roles of the 5-HT(1B)R, and its adapter protein p11, in emotional memory and object recognition memory processes by the use of p11 knockout (p11KO) mice, a genetic model for aspects of depression-related states. 5-HT(1B)R agonist treatment induced an impairing effect on emotional memory in wild type (WT) mice. In comparison, p11KO mice displayed reduced long-term emotional memory performance. Unexpectedly, 5-HT(1B)R agonist stimulation enhanced memory in p11KO mice, and this atypical switch was reversed after hippocampal adeno-associated virus mediated gene transfer of p11. Notably, 5-HT(1B)R stimulation increased glutamatergic neurotransmission in the hippocampus in p11KO mice, but not in WT mice, as measured by both pre- and postsynaptic criteria. Magnetic resonance spectroscopy demonstrated global hippocampal reductions of inhibitory GABA, which may contribute to the memory enhancement and potentiation of pre- and post-synaptic measures of glutamate transmission by a 5-HT(1B)R agonist in p11KO mice. It is concluded that the level of hippocampal p11 determines the directionality of 5-HT(1B)R action on emotional memory processing and modulates hippocampal functionality. These results emphasize the importance of using relevant disease models when evaluating the role of serotonin neurotransmission in cognitive deficits related to psychiatric disorders.
Collapse
|
49
|
The effects of congenital brain serotonin deficiency on responses to chronic fluoxetine. Transl Psychiatry 2013; 3:e291. [PMID: 23942622 PMCID: PMC3756292 DOI: 10.1038/tp.2013.65] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 07/10/2013] [Indexed: 12/17/2022] Open
Abstract
The importance of reversing brain serotonin (5-HT) deficiency and promoting hippocampal neurogenesis in the mechanisms of action for antidepressants remain highly controversial. Here we examined the behavioral, neurochemical and neurogenic effects of chronic fluoxetine (FLX) in a mouse model of congenital 5-HT deficiency, the tryptophan hydroxylase 2 (R439H) knock-in (Tph2KI) mouse. Our results demonstrate that congenital 5-HT deficiency prevents a subset of the signature molecular, cellular and behavioral effects of FLX, despite the fact that FLX restores the 5-HT levels of Tph2KI mice to essentially the levels observed in wild-type mice at baseline. These results suggest that inducing supra-physiological levels of 5-HT, not merely reversing 5-HT deficiency, is required for many of the antidepressant-like effects of FLX. We also demonstrate that co-administration of the 5-HT precursor, 5-hydroxytryptophan (5-HTP), along with FLX rescues the novelty suppressed feeding (NSF) anxiolytic-like effect of FLX in Tph2KI mice, despite still failing to induce neurogenesis. Thus, our results indicate that brain 5-HT deficiency reduces the efficacy of FLX and that supplementation with 5-HTP can restore some antidepressant-like responses in the context of 5-HT deficiency. Our findings also suggest that feeding latency reductions in the NSF induced by chronic 5-HT elevation are not mediated by drug-induced increments in neurogenesis in 5-HT-deficient animals. Overall, these findings shed new light on the impact of 5-HT deficiency on responses to FLX and may have important implications for treatment selection in depression and anxiety disorders.
Collapse
|
50
|
SMARCA3, a chromatin-remodeling factor, is required for p11-dependent antidepressant action. Cell 2013; 152:831-43. [PMID: 23415230 DOI: 10.1016/j.cell.2013.01.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 09/14/2012] [Accepted: 01/08/2013] [Indexed: 02/02/2023]
Abstract
p11, through unknown mechanisms, is required for behavioral and cellular responses to selective serotonin reuptake inhibitors (SSRIs). We show that SMARCA3, a chromatin-remodeling factor, is a target for the p11/annexin A2 heterotetrameric complex. Determination of the crystal structure indicates that SMARCA3 peptide binds to a hydrophobic pocket in the heterotetramer. Formation of this complex increases the DNA-binding affinity of SMARCA3 and its localization to the nuclear matrix fraction. In the dentate gyrus, both p11 and SMARCA3 are highly enriched in hilar mossy cells and basket cells. The SSRI fluoxetine induces expression of p11 in both cell types and increases the amount of the ternary complex of p11/annexin A2/SMARCA3. SSRI-induced neurogenesis and behavioral responses are abolished by constitutive knockout of SMARCA3. Our studies indicate a central role for a chromatin-remodeling factor in the SSRI/p11 signaling pathway and suggest an approach to the development of improved antidepressant therapies. PAPERCLIP:
Collapse
|