1
|
Zhou T, Pan J, Yan C, Yuan J, Song H, Han Y. Unveiling shared biomarkers and therapeutic targets between systemic lupus erythematosus and heart failure through bioinformatics analysis. Front Med (Lausanne) 2024; 11:1402010. [PMID: 38912340 PMCID: PMC11190381 DOI: 10.3389/fmed.2024.1402010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/21/2024] [Indexed: 06/25/2024] Open
Abstract
Background Systemic lupus erythematosus (SLE) is frequently accompanied by various complications, with cardiovascular diseases being particularly concerning due to their high mortality rate. Although there is clinical evidence suggesting a potential correlation between SLE and heart failure (HF), the underlying shared mechanism is not fully understood. Therefore, it is imperative to explore the potential mechanisms and shared therapeutic targets between SLE and HF. Methods The SLE and HF datasets were downloaded from the NCBI Gene Expression Omnibus database. Differentially expressed genes (DEGs) in both SLE and HF were performed using "limma" R package. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genes (KEGG) analyses were conducted to analyze the enriched functions and pathways of DEGs in both SLE and HF datasets. Protein-Protein Interaction network (PPI) and the molecular complex detection (MCODE) plugins in the Cytoscape software were performed to identify the shared hub genes between SLE and HF datasets. R package "limma" was utilized to validate the expression of hub genes based on SLE (GSE122459) and HF (GSE196656) datasets. CIBERSORT algorithm was utilized to analyze the immune cell infiltration of SLE and HF samples based on SLE (GSE112087) and HF (GSE116250) datasets. A weighted gene co-expression network analysis (WGCNA) network was established to further validate the hub genes based on HF dataset (GSE116250). Molecular biology techniques were conducted to validate the hub genes. Results 999 shared DGEs were identified between SLE and HF datasets, which were mainly enriched in pathways related to Th17 cell differentiation. 5 shared hub genes among the common DGEs between SLE and HF datasets were screened and validated, including HSP90AB1, NEDD8, RPLP0, UBB, and UBC. Additionally, 5 hub genes were identified in the central part of the MEbrown module, showing the strongest correlation with dilated cardiomyopathy. HSP90AB1 and UBC were upregulated in failing hearts compared to non-failing hearts, while UBB, NEDD8, and RPLP0 did not show significant changes. Conclusion HSP90AB1 and UBC are closely related to the co-pathogenesis of SLE and HF mediated by immune cell infiltration. They serve as promising molecular markers and potential therapeutic targets for the treatment of SLE combined with HF.
Collapse
Affiliation(s)
- Ting Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jing Pan
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jing Yuan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haixu Song
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yaling Han
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
2
|
Xu J, Zhou L, Chen Z, Wang Y, Xu F, Kuang Q, Zhang Y, Zheng H. Bacillus coagulans and Clostridium butyricum synergistically alleviate depression in a chronic unpredictable mild stress mouse model through altering gut microbiota and prefrontal cortex gene expression. Front Pharmacol 2024; 15:1393874. [PMID: 38855745 PMCID: PMC11158626 DOI: 10.3389/fphar.2024.1393874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/22/2024] [Indexed: 06/11/2024] Open
Abstract
Introduction: The prevalence of major depressive disorder (MDD) has gradually increased and has attracted widespread attention. The aim of this study was to investigate the effect of a probiotic compound consisting of Bacillus coagulans and Clostridium butyricum, on a mouse depression model. Methods: Mice were subjected to chronic unpredictable mild stress (CUMS) and then treated with the probiotics at different concentrations. And mice received behavior test such as forced swimming test and tail suspension test. After that, all mice were sacrificed and the samples were collected for analysis. Moreover, prefrontal cortex (PFC) gene expression and the gut microbiota among different groups were also analyzed. Results: Probiotics improved depressive-like behavior in CUMS mice, as indicated by decreased immobility time (p < 0.05) in the forced swimming test and tail suspension test. probiotics intervention also increased the level of 5-hydroxytryptamine (5-HT) in the prefrontal cortex and decreased the adrenocorticotropic hormone (ACTH) level in serum. In addition, by comparing the PFC gene expression among different groups, we found that the genes upregulated by probiotics were enriched in the PI3K-Akt signaling pathway in the prefrontal cortex. Moreover, we found that downregulated genes in prefrontal cortex of CUMS group such as Sfrp5 and Angpt2, which were correlated with depression, were reversed by the probiotics. Furthermore, the probiotics altered the structure of the gut microbiota, and reversed the reduction of cob(II)yrinate a,c-diamide biosynthesis I pathway in CUMS group. Several species like Bacteroides caecimuris and Parabacteroides distasoni, whose abundance was significantly decreased in the CUMS group but reversed after the probiotics intervention, showed significantly positive correlation with depression associated genes such as Tbxas1 and Cldn2. Discussion: These findings suggested that CUMS-induced depression-like behavior can be alleviated by the probiotics, possibly through alterations in the PFC gene expression and gut microbiota.
Collapse
Affiliation(s)
- Jingyi Xu
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Lei Zhou
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Zhaowei Chen
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Yuezhu Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Fang Xu
- The Academician Workstation, Shanghai Fourth People’s Hospital Affiliated to Tongji University, Shanghai, China
| | - Qun Kuang
- Jiangsu Limited Company of Suwei Microbiology, Wuxi, China
| | - Yixuan Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Huajun Zheng
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
- The Academician Workstation, Shanghai Fourth People’s Hospital Affiliated to Tongji University, Shanghai, China
| |
Collapse
|
3
|
White AG, Elias E, Orozco A, Robinson SA, Manners MT. Chronic Stress-Induced Neuroinflammation: Relevance of Rodent Models to Human Disease. Int J Mol Sci 2024; 25:5085. [PMID: 38791125 PMCID: PMC11121038 DOI: 10.3390/ijms25105085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
The brain is the central organ of adaptation to stress because it perceives and determines threats that induce behavioral, physiological, and molecular responses. In humans, chronic stress manifests as an enduring consistent feeling of pressure and being overwhelmed for an extended duration. This can result in a persistent proinflammatory response in the peripheral and central nervous system (CNS), resulting in cellular, physiological, and behavioral effects. Compounding stressors may increase the risk of chronic-stress-induced inflammation, which can yield serious health consequences, including mental health disorders. This review summarizes the current knowledge surrounding the neuroinflammatory response in rodent models of chronic stress-a relationship that is continually being defined. Many studies investigating the effects of chronic stress on neuroinflammation in rodent models have identified significant changes in inflammatory modulators, including nuclear factor-κB (NF-κB) and toll-like receptors (TLRs), and cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β, and IL-6. This suggests that these are key inflammatory factors in the chronic stress response, which may contribute to the establishment of anxiety and depression-like symptoms. The behavioral and neurological effects of modulating inflammatory factors through gene knockdown (KD) and knockout (KO), and conventional and alternative medicine approaches, are discussed.
Collapse
Affiliation(s)
- Abigail G. White
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA
| | - Elias Elias
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA
| | - Andrea Orozco
- Department of Psychology, Williams College, Williamstown, MA 01267, USA
| | | | - Melissa T. Manners
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA
| |
Collapse
|
4
|
Hull MA, Nunamaker EA, Reynolds PS. Effects of Refined Handling on Reproductive Indices of BALB/cJ and CD-1 IGS Mice. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2024; 63:3-9. [PMID: 38154807 PMCID: PMC10844741 DOI: 10.30802/aalas-jaalas-23-000028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/13/2023] [Accepted: 11/06/2023] [Indexed: 12/30/2023]
Abstract
Current mouse handling methods during cage change procedures can cause stress and potentially compromise animal welfare. Our previous study of breeding C57BL/6J mice found modest increases in pup production and a significant reduction in preweaning litter losses when mice were handled using a tunnel as compared with a tail-lift with padded forceps. The current study evaluated how these 2 handling methods affected reproduction by 2 additional mouse strains, BALB/cJ (a low- to intermediate-fecundity strain) and CD-1 IGS (a high-fecundity stock). We predicted that refined handling would have minimal effects on the high-fecundity line with a satisfactory production rate and greater effects on the low-fecundity line. Handling method (tunnel compared with tail-lift) was randomly assigned to monogamous breeding pairs of mice. Reproductive metrics (litter size at birth and weaning, numbers of litters, litter attrition, between-litter intervals, pup wean- ing weight, and sex ratio) were prospectively monitored for 80 BALB/cJ and 77 CD-1 pairs that were bred continuously for 6 mo. Both strains of mice were highly productive, exceeding previously published breeding data. However, neither strain demonstrated operational or statistically significant differences between handling methods for any reproduction metric. As we detected no negative effects in these 2 strains and the benefits are clear in other strains, refined handling should be considered for all breeding mice.
Collapse
Affiliation(s)
- Margaret A Hull
- Animal Care Services, University of Florida, Gainesville, Florida
| | - Elizabeth A Nunamaker
- Global Animal Welfare and Training, Charles River Laboratories, Wilmington, Massachusetts; and
| | - Penny S Reynolds
- Statistics in Anesthesiology Research Core, Department of Anesthesiology, College of Medicine, Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
5
|
Campeau S, McNulty C, Stanley JT, Gerber AN, Sasse SK, Dowell RD. Determination of steady-state transcriptome modifications associated with repeated homotypic stress in the rat rostral posterior hypothalamic region. Front Neurosci 2023; 17:1173699. [PMID: 37360161 PMCID: PMC10288150 DOI: 10.3389/fnins.2023.1173699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/18/2023] [Indexed: 06/28/2023] Open
Abstract
Chronic stress is epidemiologically correlated with physical and psychiatric disorders. Whereas many animal models of chronic stress induce symptoms of psychopathology, repeated homotypic stressors to moderate intensity stimuli typically reduce stress-related responses with fewer, if any, pathological symptoms. Recent results indicate that the rostral posterior hypothalamic (rPH) region is a significant component of the brain circuitry underlying response reductions (habituation) associated with repeated homotypic stress. To test whether posterior hypothalamic transcriptional regulation associates with the neuroendocrine modifications induced by repeated homotypic stress, RNA-seq was performed in the rPH dissected from adult male rats that experienced either no stress, 1, 3, or 7 stressful loud noise exposures. Plasma samples displayed reliable increases of corticosterone in all stressed groups, with the smallest increase in the group exposed to 7 loud noises, indicating significant habituation compared to the other stressed groups. While few or no differentially expressed genes were detected 24-h after one or three loud noise exposures, relatively large numbers of transcripts were differentially expressed between the group exposed to 7 loud noises when compared to the control or 3-stress groups, respectively, which correlated with the corticosterone response habituation observed. Gene ontology analyses indicated multiple significant functional terms related to neuron differentiation, neural membrane potential, pre- and post-synaptic elements, chemical synaptic transmission, vesicles, axon guidance and projection, glutamatergic and GABAergic neurotransmission. Some of the differentially expressed genes (Myt1l, Zmat4, Dlx6, Csrnp3) encode transcription factors that were independently predicted by transcription factor enrichment analysis to target other differentially regulated genes in this study. A similar experiment employing in situ hybridization histochemical analysis in additional animals validated the direction of change of the 5 transcripts investigated (Camk4, Gabrb2, Gad1, Grin2a and Slc32a) with a high level of temporal and regional specificity for the rPH. In aggregate, the results suggest that distinct patterns of gene regulation are obtained in response to a repeated homotypic stress regimen; they also point to a significant reorganization of the rPH region that may critically contribute to the phenotypic modifications associated with repeated homotypic stress habituation.
Collapse
Affiliation(s)
- Serge Campeau
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States
| | - Connor McNulty
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States
| | - Jacob T. Stanley
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States
- BioFrontiers Institute, University of Colorado, Boulder, CO, United States
| | - Anthony N. Gerber
- Department of Medicine, National Jewish Health, Denver, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| | - Sarah K. Sasse
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Robin D. Dowell
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States
- BioFrontiers Institute, University of Colorado, Boulder, CO, United States
- Department of Computer Science, University of Colorado, Boulder, CO, United States
| |
Collapse
|
6
|
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: 4.7] [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
|
7
|
Novak J, Jaric I, Rosso M, Rufener R, Touma C, Würbel H. Handling method affects measures of anxiety, but not chronic stress in mice. Sci Rep 2022; 12:20938. [PMID: 36463282 PMCID: PMC9719500 DOI: 10.1038/s41598-022-25090-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/24/2022] [Indexed: 12/07/2022] Open
Abstract
Studies in mice have shown that less aversive handling methods (e.g. tunnel or cup handling) can reduce behavioural measures of anxiety in comparison to picking mice up by their tail. Despite such evidence, tail handling continues to be used routinely. Besides resistance to change accustomed procedures, this may also be due to the fact that current evidence in support of less aversive handling is mostly restricted to effects of extensive daily handling, which may not apply to routine husbandry practices. The aim of our study was to assess whether, and to what extent, different handling methods during routine husbandry induce differences in behavioural and physiological measures of stress in laboratory mice. To put the effects of handling method in perspective with chronic stress, we compared handling methods to a validated paradigm of unpredictable chronic mild stress (UCMS). We housed mice of two strains (Balb/c and C57BL/6) and both sexes either under standard laboratory conditions (CTRL) or under UCMS. Half of the animals from each housing condition were tail handled and half were tunnel handled twice per week, once during a cage change and once for a routine health check. We found strain dependent effects of handling method on behavioural measures of anxiety: tunnel handled Balb/c mice interacted with the handler more than tail handled conspecifics, and tunnel handled CTRL mice showed increased open arm exploration in the elevated plus-maze. Mice undergoing UCMS showed increased plasma corticosterone levels and reduced sucrose preference. However, we found no effect of handling method on these stress-associated measures. Our results therefore indicate that routine tail handling can affect behavioural measures of anxiety, but may not be a significant source of chronic husbandry stress. Our results also highlight strain dependent responses to handling methods.
Collapse
Affiliation(s)
- Janja Novak
- grid.5734.50000 0001 0726 5157Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Ivana Jaric
- grid.5734.50000 0001 0726 5157Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Marianna Rosso
- grid.5734.50000 0001 0726 5157Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Reto Rufener
- grid.5734.50000 0001 0726 5157Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute of Parasitology, University of Bern, Bern, Switzerland
| | - Chadi Touma
- grid.10854.380000 0001 0672 4366Department of Behavioural Biology, Osnabrück University, Osnabrück, Germany
| | - Hanno Würbel
- grid.5734.50000 0001 0726 5157Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Long-Term Effects of Repeated Social Defeat Stress on Brain Activity during Social Interaction in BALB/c Mice. eNeuro 2022; 9:ENEURO.0068-22.2022. [PMID: 35437264 PMCID: PMC9070729 DOI: 10.1523/eneuro.0068-22.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/26/2022] [Accepted: 04/04/2022] [Indexed: 12/28/2022] Open
Abstract
Understanding the long-term effects of stress on brain function is crucial for understanding the mechanisms of depression. The BALB/c mouse strain has high susceptibility to stress and is thus an effective model for depression. The long-term effects of repeated social defeat stress (SDS) on BALB/c mice, however, are not clear. Here, we investigated the effects of repeated SDS in male BALB/c mice over the subsequent two weeks. Some defeated mice immediately exhibited social avoidance, whereas anxiety-like behavior was only evident at later periods. Furthermore, defeated mice segregated into two groups based on the level of social avoidance, namely, avoidant and nonavoidant mice. The characteristic of avoidance or nonavoidance in each individual was not fixed over the two weeks. In addition, we developed a semi-automated method for analyzing c-Fos expression in the mouse brain to investigate the effect of repeated SDS on brain activity more than two weeks after the end of the stress exposure. Following social interaction, c-Fos expression was reduced in several brain regions in the defeated mice compared with control mice. The correlation of c-Fos expression among these brain areas, with exception of the medial prefrontal cortex (mPFC) and central amygdala (CeA), was increased in defeated mice, suggesting increased synchrony. Notably, c-Fos expression in the lateral habenula (LHb) was different between mice that exhibited social avoidance from immediately after the repeated SDS and those that exhibited social avoidance only at later periods. These observations provide insight into the long-term effects of social stress on behavior and brain activity.
Collapse
|
9
|
Nazir S, Farooq RK, Nasir S, Hanif R, Javed A. Therapeutic effect of Thymoquinone on behavioural response to UCMS and neuroinflammation in hippocampus and amygdala in BALB/c mice model. Psychopharmacology (Berl) 2022; 239:47-58. [PMID: 35029704 DOI: 10.1007/s00213-021-06038-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 11/26/2021] [Indexed: 11/29/2022]
Abstract
RATIONALE Major depressive disorder is the leading cause of disability worldwide. The corticolimbic system plays a critical role in the emotional and cognitive aspects of major depressive disorder. Owing to the unsatisfactory efficacy of conventional antidepressants, there is a need to explore novel therapies. OBJECTIVES The current study aimed to explore the antidepressant potential of thymoquinone, a natural compound with anti-inflammatory activity, and propose its underlying mechanism of action in the unpredictable chronic mild stress (UCMS) mouse model. METHODS Coat state, forced swim test, elevated plus maze test, novelty suppressed feeding test and social interaction test were performed to quantify the behavioural shift induced by UCMS and the effect of thymoquinone and fluoxetine treatment. In addition, messenger RNA (mRNA) expression levels of inflammatory cytokines (IL-1β, IL-6 and TNF-α) and BDNF and NeuN were analysed by a quantitative real-time polymerase chain reaction in the hippocampus and amygdala of experimental and control groups. RESULTS UCMS significantly deteriorated coat state. Thymoquinone reinstated the resignation behaviour and latency to feed affected by UCMS. UCMS induced an increase in inflammatory cytokines (IL-1β, IL-6 and TNF-α) in the hippocampus and amygdala, which was decreased by thymoquinone. UCMS caused an increase in BDNF and NeuN mRNA levels in the amygdala while a decrease in the hippocampus. This opposite effect on BDNF was also compensated by thymoquinone; however, thymoquinone did not significantly change Ki67 and NeuN mRNA levels in the hippocampus. CONCLUSIONS Thymoquinone restored the behavioural changes induced by UCMS. In addition, the antidepressant effect of thymoquinone is in line with changes in inflammatory parameters and changes in BDNF in the hippocampus and amygdala.
Collapse
Affiliation(s)
- Sadia Nazir
- Atta-Ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, 44000, Pakistan
| | - Rai Khalid Farooq
- Department of Neuroscience Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Sadia Nasir
- Atta-Ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, 44000, Pakistan
| | - Rumeza Hanif
- Atta-Ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, 44000, Pakistan
| | - Aneela Javed
- Atta-Ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, 44000, Pakistan.
| |
Collapse
|
10
|
Gui S, Liu Y, Pu J, Song X, Chen X, Chen W, Zhong X, Wang H, Liu L, Xie P. Comparative analysis of hippocampal transcriptional features between major depressive disorder patients and animal models. J Affect Disord 2021; 293:19-28. [PMID: 34161882 DOI: 10.1016/j.jad.2021.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is a psychiatric disorder caused by various etiologies. Chronic stress models are used to simulate the heterogeneous pathogenic processes of depression. However, few studies have compared transcriptional features between stress models and MDD patients. METHODS We generated hippocampal transcriptional profiles of the chronic social defeat model by RNA sequencing and downloaded raw data of the same brain region from public databases of the chronic unpredictable mild stress model, the learned helplessness model, and MDD patients. Differential expression and gene co-expression analyses were integrated to compare transcriptional features between stress models and MDD patients. RESULTS Each stress model shared 11.4% to 16.3% of differentially expressed genes with MDD patients. Functional analysis at the gene expression level identified altered ensheathment of neurons in both stress models and MDD patients. At the gene network level, each stress model shared 20.9% to 41.6% of co-expressed genes with MDD patients. Functional analysis based on these genes found that axon guidance signaling is the most significantly enriched pathway that was shared by all stress models and MDD patients. LIMITATIONS This study was limited by considering only a single brain region and a single sex of stress model animals. CONCLUSIONS Our results show that hippocampal transcriptional features of stress models partially overlap with those of MDD patients. The canonical pathways of MDD patients, including ensheathment of neurons, PTEN signaling, and axonal guidance signaling, were shared with all stress models. Our findings provide further clues to understand the molecular mechanisms of depression.
Collapse
Affiliation(s)
- Siwen Gui
- College of Biomedical Engineering, Chongqing Medical University, Chongqing 40016, China; State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing 40016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yiyun Liu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Juncai Pu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xuemian Song
- College of Biomedical Engineering, Chongqing Medical University, Chongqing 40016, China; State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing 40016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaopeng Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Weiyi Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaogang Zhong
- College of Stomatology and Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Haiyang Wang
- College of Stomatology and Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Lanxiang Liu
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
11
|
White JD, Arefin TM, Pugliese A, Lee CH, Gassen J, Zhang J, Kaffman A. Early life stress causes sex-specific changes in adult fronto-limbic connectivity that differentially drive learning. eLife 2020; 9:58301. [PMID: 33259286 PMCID: PMC7725504 DOI: 10.7554/elife.58301] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 11/30/2020] [Indexed: 12/26/2022] Open
Abstract
It is currently unclear whether early life stress (ELS) affects males and females differently. However, a growing body of work has shown that sex moderates responses to stress and injury, with important insights into sex-specific mechanisms provided by work in rodents. Unfortunately, most of the ELS studies in rodents were conducted only in males, a bias that is particularly notable in translational work that has used human imaging. Here we examine the effects of unpredictable postnatal stress (UPS), a mouse model of complex ELS, using high resolution diffusion magnetic resonance imaging. We show that UPS induces several neuroanatomical alterations that were seen in both sexes and resemble those reported in humans. In contrast, exposure to UPS induced fronto-limbic hyper-connectivity in males, but either no change or hypoconnectivity in females. Moderated-mediation analysis found that these sex-specific changes are likely to alter contextual freezing behavior in males but not in females.
Collapse
Affiliation(s)
- Jordon D White
- Department of Psychiatry, Yale University School of Medicine, New Haven, United States
| | - Tanzil M Arefin
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, United States
| | - Alexa Pugliese
- Department of Psychiatry, Yale University School of Medicine, New Haven, United States
| | - Choong H Lee
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, United States
| | - Jeff Gassen
- Department of Psychology, Texas Christian University, Fort Worth, United States
| | - Jiangyang Zhang
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, United States
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, New Haven, United States
| |
Collapse
|
12
|
Wu L, Wei Y, Li H, Li W, Gu C, Sun J, Xia H, Zhang J, Chen F, Liu Q. The ubiquitination and acetylation of histones are associated with male reproductive disorders induced by chronic exposure to arsenite. Toxicol Appl Pharmacol 2020; 408:115253. [PMID: 32991915 DOI: 10.1016/j.taap.2020.115253] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022]
Abstract
Exposure to arsenic, which occurs via various routes, can cause reproductive toxicity. However, the mechanism for arsenic-induced reproductive disorders in male mice has not been extensively investigated. Here, 6-week-old male mice were dosed to 0, 5, 10, or 20 ppm sodium arsenite (NaAsO2), an active form of arsenic, in drinking water for six months. For male mice exposed to arsenite, fertility was lower compared to control mice. Moreover, for exposed mice, there were lower sperm counts, lower sperm motility, and higher sperm malformation ratios. Further, the mRNA and protein levels of the gonadotropin-regulated testicular RNA helicase (DDX25) and chromosome region maintenance-1 protein (CRM1), along with proteins associated with high mobility group box 2 (HMGB2), phosphoglycerate kinase 2 (PGK2), and testicular angiotensin-converting enzyme (tACE) were lower. Furthermore, chronic exposure to arsenite led to lower H2A ubiquitination (ubH2A); histone H3 acetylation K18 (H3AcK18); and histone H4 acetylations K5, K8, K12, and K16 (H4tetraAck) in haploid spermatids from testicular tissues. These alterations disrupted deposition of protamine 1 (Prm1) in testes. Overall, the present results indicate that the ubiquitination and acetylation of histones is involved in the spermiogenesis disorders caused by chronic exposure to arsenite, which points to a previously unknown connection between the modification of histones and arsenite-induced male reproductive toxicity.
Collapse
Affiliation(s)
- Lu Wu
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Yongyue Wei
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Han Li
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Wenqi Li
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Chenxi Gu
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Jing Sun
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Haibo Xia
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Jingshu Zhang
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; Jiangsu Safety Assessment and Research Center for Drug, Pesticide, and Veterinary Drug, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Feng Chen
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.
| | - Qizhan Liu
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.
| |
Collapse
|
13
|
Musaelyan K, Yildizoglu S, Bozeman J, Du Preez A, Egeland M, Zunszain PA, Pariante CM, Fernandes C, Thuret S. Chronic stress induces significant gene expression changes in the prefrontal cortex alongside alterations in adult hippocampal neurogenesis. Brain Commun 2020; 2:fcaa153. [PMID: 33543135 PMCID: PMC7850288 DOI: 10.1093/braincomms/fcaa153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 07/20/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
Adult hippocampal neurogenesis is involved in stress-related disorders such as depression, posttraumatic stress disorders, as well as in the mechanism of antidepressant effects. However, the molecular mechanisms involved in these associations remain to be fully explored. In this study, unpredictable chronic mild stress in mice resulted in a deficit in neuronal dendritic tree development and neuroblast migration in the hippocampal neurogenic niche. To investigate molecular pathways underlying neurogenesis alteration, genome-wide gene expression changes were assessed in the prefrontal cortex, hippocampus and the hypothalamus alongside neurogenesis changes. Cluster analysis showed that the transcriptomic signature of chronic stress is much more prominent in the prefrontal cortex compared to the hippocampus and the hypothalamus. Pathway analyses suggested huntingtin, leptin, myelin regulatory factor, methyl-CpG binding protein and brain-derived neurotrophic factor as the top predicted upstream regulators of transcriptomic changes in the prefrontal cortex. Involvement of the satiety regulating pathways (leptin) was corroborated by behavioural data showing increased food reward motivation in stressed mice. Behavioural and gene expression data also suggested circadian rhythm disruption and activation of circadian clock genes such as Period 2. Interestingly, most of these pathways have been previously shown to be involved in the regulation of adult hippocampal neurogenesis. It is possible that activation of these pathways in the prefrontal cortex by chronic stress indirectly affects neuronal differentiation and migration in the hippocampal neurogenic niche via reciprocal connections between the two brain areas.
Collapse
Affiliation(s)
- Ksenia Musaelyan
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Selin Yildizoglu
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| | - James Bozeman
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| | - Andrea Du Preez
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Martin Egeland
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Patricia A Zunszain
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| | - Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| | - Cathy Fernandes
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London SE1 1UL, UK
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| |
Collapse
|
14
|
Warden AS, DaCosta A, Mason S, Blednov YA, Mayfield RD, Harris RA. Inbred Substrain Differences Influence Neuroimmune Response and Drinking Behavior. Alcohol Clin Exp Res 2020; 44:1760-1768. [PMID: 32640038 DOI: 10.1111/acer.14410] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/03/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND The inbred mouse strain C57BL/6 is widely used in both models of addiction and immunological disease. However, there are pronounced phenotypic differences in ethanol (EtOH) consumption and innate immune response between C57BL/6 substrains. The focus of this study was to examine the effects of substrain on innate immune response and neuroimmune-induced escalation of voluntary EtOH consumption. The main goal was to identify whether substrain differences in immune response can account for differences in EtOH behavior. METHODS We compared acute innate immune response with a viral dsRNA mimic, polyinosinic:polycytidylic acid (poly(I:C)), in brain using qRT-PCR in both C57BL/6N and C57BL/6J mice. Next, we used a neuroimmune model of escalation using poly(I:C) to compare drinking behavior between substrains. Finally, we compared brain neuroimmune response with both EtOH and repeated poly(I:C) in both substrains as a way to account for differences in EtOH behavior. RESULTS We found that C57BL/6 substrains have differing immune response and drinking behaviors. C57BL/6N mice have a shorter but more robust inflammatory response to acute poly(I:C). In contrast, C57BL/6J mice have a smaller but longer-lasting acute immune response to poly(I:C). In our neuroimmune-induced escalation model, C57BL/6J mice but not C57BL/6N mice escalate EtOH intake after poly(I:C). Finally, only C57BL/6J mice show enhanced proinflammatory transcript abundance after poly(I:C) and EtOH, suggesting that longer-lasting immune responses are critical to neuroimmune drinking phenotypes. CONCLUSIONS Altogether, this work has elucidated additional influences that substrain has on both innate immune response and drinking phenotypes. Our observations highlight the importance of considering and reporting the source and background used for production of transgenic and knockout mice. These data provide further evidence that genetic background must be carefully considered when investigating the role of neuroimmune signaling in EtOH abuse.
Collapse
Affiliation(s)
- Anna S Warden
- From the Waggoner Center for Alcoholism and Addiction Research, The University of Texas at Austin, Austin, Texas, USA.,Institute for Neuroscience, The University of Texas at Austin, Austin, Texas, USA.,Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA
| | - Adriana DaCosta
- From the Waggoner Center for Alcoholism and Addiction Research, The University of Texas at Austin, Austin, Texas, USA
| | - Sonia Mason
- From the Waggoner Center for Alcoholism and Addiction Research, The University of Texas at Austin, Austin, Texas, USA
| | - Yuri A Blednov
- From the Waggoner Center for Alcoholism and Addiction Research, The University of Texas at Austin, Austin, Texas, USA
| | - Roy Dayne Mayfield
- From the Waggoner Center for Alcoholism and Addiction Research, The University of Texas at Austin, Austin, Texas, USA.,Institute for Neuroscience, The University of Texas at Austin, Austin, Texas, USA
| | - Robert Adron Harris
- From the Waggoner Center for Alcoholism and Addiction Research, The University of Texas at Austin, Austin, Texas, USA.,Institute for Neuroscience, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
15
|
Scarpa JR, Fatma M, Loh YHE, Traore SR, Stefan T, Chen TH, Nestler EJ, Labonté B. Shared Transcriptional Signatures in Major Depressive Disorder and Mouse Chronic Stress Models. Biol Psychiatry 2020; 88:159-168. [PMID: 32169281 PMCID: PMC7740570 DOI: 10.1016/j.biopsych.2019.12.029] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/03/2019] [Accepted: 12/24/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND Most of our knowledge of the biological basis of major depressive disorder (MDD) is derived from studies of chronic stress models in rodents. While these models capture certain aspects of the behavioral and neuroendocrine features of MDD, the extent to which they reproduce the molecular pathology of the human syndrome remains unknown. METHODS We systematically compared transcriptional signatures in two brain regions implicated in depression-medial prefrontal cortex and nucleus accumbens-of humans with MDD and of 3 chronic stress models in mice: chronic variable stress, adult social isolation, and chronic social defeat stress. We used differential expression analysis combined with weighted gene coexpression network analysis to create interspecies gene networks and assess the capacity of each stress paradigm to recapitulate the transcriptional organization of gene networks in human MDD. RESULTS Our results show significant overlap between transcriptional alterations in medial prefrontal cortex and nucleus accumbens in human MDD and the 3 mouse chronic stress models, with each of the chronic stress paradigms capturing distinct aspects of MDD abnormalities. Chronic variable stress and adult social isolation better reproduce differentially expressed genes, while chronic social defeat stress and adult social isolation better reproduce gene networks characteristic of human MDD. We also identified several gene networks and their constituent genes that are most significantly associated with human MDD and mouse stress models. CONCLUSIONS This study demonstrates the ability of 3 chronic stress models in mice to recapitulate distinct aspects of the broad molecular pathology of human MDD, with no one mouse model apparently better than another.
Collapse
Affiliation(s)
- Joseph R Scarpa
- Department of Anesthesiology, Weill Cornell Medicine, New York, New York
| | - Mena Fatma
- Department of Psychiatry and Neurosciences, Laval University, Québec, Québec, Canada
| | - Yong-Hwee E Loh
- Norris Medical Library, University of Southern California, Los Angeles, California
| | - Said Romaric Traore
- Department of Psychiatry and Neurosciences, Laval University, Québec, Québec, Canada
| | - Theo Stefan
- Department of Psychiatry and Neurosciences, Laval University, Québec, Québec, Canada
| | - Ting Huei Chen
- Department of Psychiatry and Neurosciences, Laval University, Québec, Québec, Canada
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Benoit Labonté
- Department of Psychiatry and Neurosciences, Laval University, Québec, Québec, Canada.
| |
Collapse
|
16
|
The effect of high fat, high sugar, and combined high fat-high sugar diets on spatial learning and memory in rodents: A meta-analysis. Neurosci Biobehav Rev 2019; 107:399-421. [DOI: 10.1016/j.neubiorev.2019.08.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 08/02/2019] [Accepted: 08/12/2019] [Indexed: 12/29/2022]
|
17
|
Misiewicz Z, Iurato S, Kulesskaya N, Salminen L, Rodrigues L, Maccarrone G, Martins J, Czamara D, Laine MA, Sokolowska E, Trontti K, Rewerts C, Novak B, Volk N, Park DI, Jokitalo E, Paulin L, Auvinen P, Voikar V, Chen A, Erhardt A, Turck CW, Hovatta I. Multi-omics analysis identifies mitochondrial pathways associated with anxiety-related behavior. PLoS Genet 2019; 15:e1008358. [PMID: 31557158 PMCID: PMC6762065 DOI: 10.1371/journal.pgen.1008358] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/08/2019] [Indexed: 01/10/2023] Open
Abstract
Stressful life events are major environmental risk factors for anxiety disorders, although not all individuals exposed to stress develop clinical anxiety. The molecular mechanisms underlying the influence of environmental effects on anxiety are largely unknown. To identify biological pathways mediating stress-related anxiety and resilience to it, we used the chronic social defeat stress (CSDS) paradigm in male mice of two inbred strains, C57BL/6NCrl (B6) and DBA/2NCrl (D2), that differ in their susceptibility to stress. Using a multi-omics approach, we identified differential mRNA, miRNA and protein expression changes in the bed nucleus of the stria terminalis (BNST) and blood cells after chronic stress. Integrative gene set enrichment analysis revealed enrichment of mitochondrial-related genes in the BNST and blood of stressed mice. To translate these results to human anxiety, we investigated blood gene expression changes associated with exposure-induced panic attacks. Remarkably, we found reduced expression of mitochondrial-related genes in D2 stress-susceptible mice and in exposure-induced panic attacks in humans, but increased expression of these genes in B6 stress-susceptible mice. Moreover, stress-susceptible vs. stress-resilient B6 mice displayed more mitochondrial cross-sections in the post-synaptic compartment after CSDS. Our findings demonstrate mitochondrial-related alterations in gene expression as an evolutionarily conserved response in stress-related behaviors and validate the use of cross-species approaches in investigating the biological mechanisms underlying anxiety disorders.
Collapse
Affiliation(s)
- Zuzanna Misiewicz
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychology and Logopedics, Medicum, University of Helsinki, Helsinki, Finland
| | - Stella Iurato
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Natalia Kulesskaya
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
- Department of Psychology and Logopedics, Medicum, University of Helsinki, Helsinki, Finland
| | - Laura Salminen
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Luis Rodrigues
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Giuseppina Maccarrone
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jade Martins
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Darina Czamara
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Mikaela A. Laine
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
- Department of Psychology and Logopedics, Medicum, University of Helsinki, Helsinki, Finland
| | - Ewa Sokolowska
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Kalevi Trontti
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
- Department of Psychology and Logopedics, Medicum, University of Helsinki, Helsinki, Finland
| | - Christiane Rewerts
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Bozidar Novak
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Naama Volk
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Dong Ik Park
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Eija Jokitalo
- Electron Microscopy Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Lars Paulin
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Petri Auvinen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Vootele Voikar
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Angelika Erhardt
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
- * E-mail: (AE); (CWT); (IH)
| | - Christoph W. Turck
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
- * E-mail: (AE); (CWT); (IH)
| | - Iiris Hovatta
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
- Department of Psychology and Logopedics, Medicum, University of Helsinki, Helsinki, Finland
- * E-mail: (AE); (CWT); (IH)
| |
Collapse
|
18
|
McWhirt J, Sathyanesan M, Sampath D, Newton SS. Effects of restraint stress on the regulation of hippocampal glutamate receptor and inflammation genes in female C57BL/6 and BALB/c mice. Neurobiol Stress 2019; 10:100169. [PMID: 31193545 PMCID: PMC6535649 DOI: 10.1016/j.ynstr.2019.100169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/10/2019] [Accepted: 04/30/2019] [Indexed: 12/18/2022] Open
Abstract
The two strains of inbred mice, BALB/c and C57BL/6, are widely used in pre-clinical psychiatry research due to their differences in stress susceptibility. Gene profiling studies in these strains have implicated the inflammation pathway as the main contributor to these differences. We focused our attention on female mice and tested their response to 5- or 10-day exposure to restraint stress. We examined the stress induced changes in the regulation of 11 inflammatory cytokine genes and 12 glutamate receptor genes in the hippocampus of female BALB/c and C57BL/6 mice using quantitative PCR. Elevated proinflammatory cytokine genes include Tumor Necrosis Factor alpha (TNFa), nuclear factor kappa-light-chain-enhancer of activated B cells (NFKB), Interleukin 1 alpha (IL1a), Interleukin 1 receptor (IL1R), Interleukin 10 receptor alpha subunit (IL10Ra), Interleukin 10 receptor beta subunit (IL10Rb), and tumor necrosis factor (TNF) super family members. Our results show that BALB/c and C57BL/6 mice differ in the genes induced in response to stress exposure and the level of gene regulation change. Our results show that the gene regulation in female BALB/c and C57BL/6 mice differs between strains in the genes regulated and the magnitude of the changes.
Collapse
Affiliation(s)
- Joshua McWhirt
- Division of Basic Biomedical Sciences, University of South Dakota, Vermillion, SD, 57069, USA
| | - Monica Sathyanesan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, Sioux Falls VA Healthcare System, Sioux Falls, SD, 57105, USA
| | - Dayalan Sampath
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Samuel S Newton
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, Sioux Falls VA Healthcare System, Sioux Falls, SD, 57105, USA
| |
Collapse
|
19
|
Antoniuk S, Bijata M, Ponimaskin E, Wlodarczyk J. Chronic unpredictable mild stress for modeling depression in rodents: Meta-analysis of model reliability. Neurosci Biobehav Rev 2019; 99:101-116. [DOI: 10.1016/j.neubiorev.2018.12.002] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 12/03/2018] [Accepted: 12/03/2018] [Indexed: 01/01/2023]
|
20
|
REM sleep's unique associations with corticosterone regulation, apoptotic pathways, and behavior in chronic stress in mice. Proc Natl Acad Sci U S A 2019; 116:2733-2742. [PMID: 30683720 PMCID: PMC6377491 DOI: 10.1073/pnas.1816456116] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sleep disturbances are common in stress-related disorders but the nature of these sleep disturbances and how they relate to changes in the stress hormone corticosterone and changes in gene expression remained unknown. Here we demonstrate that in response to chronic mild stress, rapid–eye-movement sleep (REMS), a sleep state involved in emotion regulation and fear conditioning, changed first and more so than any other measured sleep characteristic. Transcriptomic profiles related to REMS continuity and theta oscillations overlapped with those for corticosterone, as well as with predictors for anhedonia, and were enriched for apoptotic pathways. These data highlight the central role of REMS in response to stress and warrant further investigation into REMS’s involvement in stress-related mental health disorders. One of sleep’s putative functions is mediation of adaptation to waking experiences. Chronic stress is a common waking experience; however, which specific aspect of sleep is most responsive, and how sleep changes relate to behavioral disturbances and molecular correlates remain unknown. We quantified sleep, physical, endocrine, and behavioral variables, as well as the brain and blood transcriptome in mice exposed to 9 weeks of unpredictable chronic mild stress (UCMS). Comparing 46 phenotypic variables revealed that rapid–eye-movement sleep (REMS), corticosterone regulation, and coat state were most responsive to UCMS. REMS theta oscillations were enhanced, whereas delta oscillations in non-REMS were unaffected. Transcripts affected by UCMS in the prefrontal cortex, hippocampus, hypothalamus, and blood were associated with inflammatory and immune responses. A machine-learning approach controlling for unspecific UCMS effects identified transcriptomic predictor sets for REMS parameters that were enriched in 193 pathways, including some involved in stem cells, immune response, and apoptosis and survival. Only three pathways were enriched in predictor sets for non-REMS. Transcriptomic predictor sets for variation in REMS continuity and theta activity shared many pathways with corticosterone regulation, in particular pathways implicated in apoptosis and survival, including mitochondrial apoptotic machinery. Predictor sets for REMS and anhedonia shared pathways involved in oxidative stress, cell proliferation, and apoptosis. These data identify REMS as a core and early element of the response to chronic stress, and identify apoptosis and survival pathways as a putative mechanism by which REMS may mediate the response to stressful waking experiences.
Collapse
|
21
|
Araujo SM, Poetini MR, Bortolotto VC, de Freitas Couto S, Pinheiro FC, Meichtry LB, de Almeida FP, Santos Musachio EA, de Paula MT, Prigol M. Chronic unpredictable mild stress-induced depressive-like behavior and dysregulation of brain levels of biogenic amines in Drosophila melanogaster. Behav Brain Res 2018; 351:104-113. [DOI: 10.1016/j.bbr.2018.05.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/09/2018] [Accepted: 05/15/2018] [Indexed: 12/16/2022]
|
22
|
Genetic Control of Myelin Plasticity after Chronic Psychosocial Stress. eNeuro 2018; 5:eN-NWR-0166-18. [PMID: 30073192 PMCID: PMC6071195 DOI: 10.1523/eneuro.0166-18.2018] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/13/2018] [Accepted: 06/25/2018] [Indexed: 12/19/2022] Open
Abstract
Anxiety disorders often manifest in genetically susceptible individuals after psychosocial stress, but the mechanisms underlying these gene-environment interactions are largely unknown. We used the chronic social defeat stress (CSDS) mouse model to study resilience and susceptibility to chronic psychosocial stress. We identified a strong genetic background effect in CSDS-induced social avoidance (SA) using four inbred mouse strains: 69% of C57BL/6NCrl (B6), 23% of BALB/cAnNCrl, 19% of 129S2/SvPasCrl, and 5% of DBA/2NCrl (D2) mice were stress resilient. Furthermore, different inbred mouse strains responded differently to stress, suggesting they use distinct coping strategies. To identify biological pathways affected by CSDS, we used RNA-sequencing (RNA-seq) of three brain regions of two strains, B6 and D2: medial prefrontal cortex (mPFC), ventral hippocampus (vHPC), and bed nucleus of the stria terminalis (BNST). We discovered overrepresentation of oligodendrocyte (OLG)-related genes in the differentially expressed gene population. Because OLGs myelinate axons, we measured myelin thickness and found significant region and strain-specific differences. For example, in resilient D2 mice, mPFC axons had thinner myelin than controls, whereas susceptible B6 mice had thinner myelin than controls in the vHPC. Neither myelin-related gene expression in several other regions nor corpus callosum thickness differed between stressed and control animals. Our unbiased gene expression experiment suggests that myelin plasticity is a substantial response to chronic psychosocial stress, varies across brain regions, and is genetically controlled. Identification of genetic regulators of the myelin response will provide mechanistic insight into the molecular basis of stress-related diseases, such as anxiety disorders, a critical step in developing targeted therapy.
Collapse
|
23
|
Silva CP, Horton WJ, Caruso MJ, Sebastian A, Klein LC, Albert I, Kamens HM. The influence of adolescent nicotine exposure on ethanol intake and brain gene expression. PLoS One 2018; 13:e0198935. [PMID: 29912970 PMCID: PMC6005571 DOI: 10.1371/journal.pone.0198935] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/29/2018] [Indexed: 01/14/2023] Open
Abstract
Nicotine and alcohol are often co-abused. Adolescence is a vulnerable period for the initiation of both nicotine and alcohol use, which can lead to subsequent neurodevelopmental and behavioral alterations. It is possible that during this vulnerable period, use of one drug leads to neurobiological alterations that affect subsequent consumption of the other drug. The aim of the present study was to determine the effect of nicotine exposure during adolescence on ethanol intake, and the effect of these substances on brain gene expression. Forty-three adolescent female C57BL/6J mice were assigned to four groups. In the first phase of the experiment, adolescent mice (PND 36-41 days) were exposed to three bottles filled with water or nicotine (200 μg/ml) for 22 h a day and a single bottle of water 2 h a day for six days. In the second phase (PND 42-45 days), the 4-day Drinking-in-the-Dark paradigm consisting of access to 20% v/v ethanol or water for 2h or 4h (the last day) was overlaid during the time when the mice did not have nicotine available. Ethanol consumption (g/kg) and blood ethanol concentrations (BEC, mg %) were measured on the final day and whole brains including the cerebellum, were dissected for RNA sequencing. Differentially expressed genes (DEG) were detected with CuffDiff and gene networks were built using WGCNA. Prior nicotine exposure increased ethanol consumption and resulting BEC. Significant DEG and biological pathways found in the group exposed to both nicotine and ethanol included genes important in stress-related neuropeptide signaling, hypothalamic-pituitary-adrenal (HPA) axis activity, glutamate release, GABA signaling, and dopamine release. These results replicate our earlier findings that nicotine exposure during adolescence increases ethanol consumption and extends this work by examining gene expression differences which could mediate these behavioral effects.
Collapse
Affiliation(s)
- Constanza P. Silva
- Biobehavioral Health Department, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - William J. Horton
- Department of Animal Science, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Michael J. Caruso
- Biobehavioral Health Department, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Aswathy Sebastian
- Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Laura C. Klein
- Biobehavioral Health Department, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Istvan Albert
- Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Helen M. Kamens
- Biobehavioral Health Department, Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
24
|
Gosselin T, Le Guisquet AM, Brizard B, Hommet C, Minier F, Belzung C. Fluoxetine induces paradoxical effects in C57BL6/J mice: comparison with BALB/c mice. Behav Pharmacol 2018; 28:466-476. [PMID: 28609327 DOI: 10.1097/fbp.0000000000000321] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The C57BL6/J mouse is the most commonly used strain in genetic investigations and behavioural tests. However, only a few studies have used C57BL6/J mice to assess the effects of antidepressant compounds. We carried out a study to compare the behavioural effects of fluoxetine (FLX) in a model of depression in two mice strains: C57BL6/J and BALB/c. We used an 8-week unpredictable chronic mild stress (UCMS) protocol during which FLX was administered (15 mg/kg, oral) from the third week to the end of the protocol. We found that UCMS induced degradation of the coat state in the two strains. Moreover, as expected, we observed that FLX elicited antidepressant-like effects in the BALB/c mice by reducing the coat state deterioration and the latency of grooming in splash test. However, in the C57BL6/J mice, it did not induce this action, but instead triggered an opposite effect: an increased sniffing latency in the novelty suppression of feeding test. We conclude that FLX exerts a paradoxical effect in the C57Bl6/J strain. This observation is consistent with some clinical features of hyper-reactivity to FLX observed in humans. Therefore, the UCMS protocol used in C57Bl6/J mice could be a good model to study the mechanisms of the paradoxical effects caused by selective serotonin reuptake inhibitors.
Collapse
Affiliation(s)
- Thomas Gosselin
- INSERM U930, Team 'Affective disorders', University of François Rabelais, Tours, France
| | | | | | | | | | | |
Collapse
|
25
|
Laviola G, Zoratto F, Ingiosi D, Carito V, Huzard D, Fiore M, Macrì S. Low empathy-like behaviour in male mice associates with impaired sociability, emotional memory, physiological stress reactivity and variations in neurobiological regulations. PLoS One 2017; 12:e0188907. [PMID: 29200428 PMCID: PMC5714342 DOI: 10.1371/journal.pone.0188907] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 11/15/2017] [Indexed: 12/30/2022] Open
Abstract
Deficits in empathy have been proposed to constitute a hallmark of several psychiatric disturbances like conduct disorder, antisocial and narcissistic personality disorders. Limited sensitivity to punishment, shallow or deficient affect and reduced physiological reactivity to environmental stressors have been often reported to co-occur with limited empathy and contribute to the onset of antisocial phenotypes. Empathy in its simplest form (i.e. emotional contagion) is addressed in preclinical models through the evaluation of the social transmission of emotional states: mice exposed to a painful stimulus display a higher response if in the presence of a familiar individual experiencing a higher degree of discomfort, than in isolation. In the present study, we investigated whether a reduction of emotional contagion can be considered a predictor of reduced sociality, sensitivity to punishment and physiological stress reactivity. To this aim, we first evaluated emotional contagion in a group of Balb/cJ mice and then discretised their values in four quartiles. The upper (i.e. Emotional Contagion Prone, ECP) and the lower (i.e. Emotional Contagion Resistant, ECR) quartiles constituted the experimental groups. Our results indicate that mice in the lower quartile are characterized by reduced sociability, impaired memory of negative events and dampened hypothalamic-pituitary-adrenocortical reactivity to external stressors. Furthermore, in the absence of changes in oxytocin receptor density, we show that these mice exhibit elevated concentrations of oxytocin and vasopressin and reduced density of BDNF receptors in behaviourally-relevant brain areas. Thus, not only do present results translate to the preclinical investigation of psychiatric disturbances, but also they can contribute to the study of emotional contagion in terms of its adaptive significance.
Collapse
Affiliation(s)
- Giovanni Laviola
- Reference Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità (ISS), Rome, Italy
- * E-mail:
| | - Francesca Zoratto
- Reference Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Danilo Ingiosi
- Reference Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Valentina Carito
- Institute of Cell Biology and Neurobiology, National Research Council of Italy (CNR), Rome, Italy
| | - Damien Huzard
- Laboratory of Behavioural Genetics, Brain Mind Institute, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Marco Fiore
- Institute of Cell Biology and Neurobiology, National Research Council of Italy (CNR), Rome, Italy
| | - Simone Macrì
- Reference Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità (ISS), Rome, Italy
| |
Collapse
|
26
|
Labonté B, Engmann O, Purushothaman I, Menard C, Wang J, Tan C, Scarpa JR, Moy G, Loh YHE, Cahill M, Lorsch ZS, Hamilton PJ, Calipari ES, Hodes GE, Issler O, Kronman H, Pfau M, Obradovic ALJ, Dong Y, Neve RL, Russo S, Kazarskis A, Tamminga C, Mechawar N, Turecki G, Zhang B, Shen L, Nestler EJ. Sex-specific transcriptional signatures in human depression. Nat Med 2017; 23:1102-1111. [PMID: 28825715 DOI: 10.1038/nm.4386] [Citation(s) in RCA: 505] [Impact Index Per Article: 63.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 07/17/2017] [Indexed: 02/08/2023]
Abstract
Major depressive disorder (MDD) is a leading cause of disease burden worldwide. While the incidence, symptoms and treatment of MDD all point toward major sex differences, the molecular mechanisms underlying this sexual dimorphism remain largely unknown. Here, combining differential expression and gene coexpression network analyses, we provide a comprehensive characterization of male and female transcriptional profiles associated with MDD across six brain regions. We overlap our human profiles with those from a mouse model, chronic variable stress, and capitalize on converging pathways to define molecular and physiological mechanisms underlying the expression of stress susceptibility in males and females. Our results show a major rearrangement of transcriptional patterns in MDD, with limited overlap between males and females, an effect seen in both depressed humans and stressed mice. We identify key regulators of sex-specific gene networks underlying MDD and confirm their sex-specific impact as mediators of stress susceptibility. For example, downregulation of the female-specific hub gene Dusp6 in mouse prefrontal cortex mimicked stress susceptibility in females, but not males, by increasing ERK signaling and pyramidal neuron excitability. Such Dusp6 downregulation also recapitulated the transcriptional remodeling that occurs in prefrontal cortex of depressed females. Together our findings reveal marked sexual dimorphism at the transcriptional level in MDD and highlight the importance of studying sex-specific treatments for this disorder.
Collapse
Affiliation(s)
- Benoit Labonté
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Olivia Engmann
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Immanuel Purushothaman
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Caroline Menard
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Junshi Wang
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Chunfeng Tan
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joseph R Scarpa
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gregory Moy
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yong-Hwee E Loh
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael Cahill
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Zachary S Lorsch
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Peter J Hamilton
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Erin S Calipari
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Georgia E Hodes
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Orna Issler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hope Kronman
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Madeline Pfau
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aleksandar L J Obradovic
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rachael L Neve
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Scott Russo
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Andrew Kazarskis
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Carol Tamminga
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Naguib Mechawar
- Department of Psychiatry, McGill University, Montreal, Québec, Canada.,McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, Québec, Canada
| | - Gustavo Turecki
- Department of Psychiatry, McGill University, Montreal, Québec, Canada.,McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, Québec, Canada
| | - Bin Zhang
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Li Shen
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Eric J Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
27
|
Wang J, Liu Y, Li L, Qi Y, Zhang Y, Li L, Teng L, Wang D. Dopamine and serotonin contribute to Paecilomyces hepiali against chronic unpredictable mild stress induced depressive behavior in Sprague Dawley rats. Mol Med Rep 2017; 16:5675-5682. [DOI: 10.3892/mmr.2017.7261] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 06/21/2017] [Indexed: 11/06/2022] Open
|
28
|
Jiang MD, Zheng Y, Wang JL, Wang YF. Drug induces depression-like phenotypes and alters gene expression profiles in Drosophila. Brain Res Bull 2017. [PMID: 28625786 DOI: 10.1016/j.brainresbull.2017.06.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND Major depressive disorder (MDD) is a severe mental illness that affects more than 350 million people worldwide. However, the molecular mechanisms of depression are currently unclear. Studies suggest that Drosophila and humans have similar depression-like symptoms under pressure. In this research, we choose Drosophila melanogaster as the animal model to explore the molecular mechanisms that trigger depression. RESULTS We found that feeding D. melanogaster with the medium containing Levodopa or Chlorpromazine could induce depression-like phenotypes in both behavioral and biochemical biomarkers, including significantly decreased food intake, mating frequency, serotonin (5-HT) concentration, and increased malondialdehyde (MDA) concentration as well as reduced activity of superoxide dismutase (SOD). Moreover, the progeny of Chlorpromazine-treated flies also showed these depression-like features. By RNA-seq technology, we identified 467 genes that were differentially expressed between Chlorpromazine treated (CPZ) and control male flies [fold-change of ≥2 (q-value<5%)]. When comparing CPZ with control flies, 312 genes were upregulated and 155 genes downregulated. Differential expression of genes related to metabolic pathway, Parkinson's disease, Huntington's disease, Alzheimer's disease and lysozyme pathways were observed. Quantitative reverse transcriptase PCR (qRT-PCR) confirmed that 19 genes are differentially expressed in CPZ and control male flies. CONCLUSIONS Levodopa, or Chlorpromazine can induce depression-like phenotypes in D. melanogaster regarding changes of appetite and sexual activity, and some key biochemical markers. A total of 467 genes were identified by RNA-seq analysis to have at least a 2-fold-change in expression between CPZ and control flies, including genes involved in metabolism, neurological diseases and lysozyme pathways. Our data provide additional insight into molecular mechanisms underlying depressive disorders in humans and may also contribute to clinical treatment.
Collapse
Affiliation(s)
- Ming-Di Jiang
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, 430079, China.
| | - Ya Zheng
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, 430079, China.
| | - Jia-Lin Wang
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, 430079, China.
| | - Yu-Feng Wang
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, 430079, China.
| |
Collapse
|
29
|
Sathyanesan M, Haiar JM, Watt MJ, Newton SS. Restraint stress differentially regulates inflammation and glutamate receptor gene expression in the hippocampus of C57BL/6 and BALB/c mice. Stress 2017; 20:197-204. [PMID: 28274152 PMCID: PMC5724770 DOI: 10.1080/10253890.2017.1298587] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The inbred mouse strains, C57BL/6 and BALB/c have been used widely in preclinical psychiatric research. The differences in stress susceptibility of available strains has provided a useful platform to test pharmacological agents and behavioral responses. Previous brain gene profiling efforts have indicated that the inflammation and immune response gene pathway is the predominant gene network in the differential stress response of BALB/c and C57BL/6 mice. The implication is that a composite stress paradigm that includes a sequence of extended, varied and unpredictable stressors induces inflammation-related genes in the hippocampus. We hypothesized that the regulation of inflammation genes in the brain could constitute a primary stress response and tested this by employing a simple stress protocol, repeated exposure to the same stressor for 10 days, 2 h of restraint per day. We examined stress-induced regulation of 13 proinflammatory cytokine genes in male BALB/c and C57BL/6 mice using quantitative PCR. Elevated cytokine genes included tumor necrosis factor alpha (TNFα), interleukin 6 (IL6), interleukin 10 (IL10), tumor necrosis factor (TNF) super family members and interleukin 1 receptor 1 (IL1R1). In addition, we examined restraint stress-induced regulation of 12 glutamate receptor genes in both strains. Our results show that restraint stress is sufficient to elevate the expression of inflammation-related genes in the hippocampus of both BABLB/c and C57BL/6 mice, but they differ in the genes that are induced and the magnitude of change. Cell types that are involved in this response include endothelial cells and astrocytes. Lay summary Repeated exposure to a simple restraint stress altered the activities of genes involved in inflammation and the functions of the excitatory neurotransmitter, glutamate. These changes in the hippocampus of the mouse brain showed differences that were dependent on the strain of mice and the length of the stress exposure. The effects of stress on activity of these genes may lead to alterations in behavior.
Collapse
Affiliation(s)
- Monica Sathyanesan
- a Division of Basic Biomedical Sciences , Sanford School of Medicine, University of South Dakota , Vermillion , SD , USA
| | - Jacob M Haiar
- a Division of Basic Biomedical Sciences , Sanford School of Medicine, University of South Dakota , Vermillion , SD , USA
| | - Michael J Watt
- a Division of Basic Biomedical Sciences , Sanford School of Medicine, University of South Dakota , Vermillion , SD , USA
| | - Samuel S Newton
- a Division of Basic Biomedical Sciences , Sanford School of Medicine, University of South Dakota , Vermillion , SD , USA
| |
Collapse
|
30
|
How age, sex and genotype shape the stress response. Neurobiol Stress 2016; 6:44-56. [PMID: 28229108 PMCID: PMC5314441 DOI: 10.1016/j.ynstr.2016.11.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/19/2016] [Accepted: 11/21/2016] [Indexed: 12/21/2022] Open
Abstract
Exposure to chronic stress is a leading pre-disposing factor for several neuropsychiatric disorders as it often leads to maladaptive responses. The response to stressful events is heterogeneous, underpinning a wide spectrum of distinct changes amongst stress-exposed individuals'. Several factors can underlie a different perception to stressors and the setting of distinct coping strategies that will lead to individual differences on the susceptibility/resistance to stress. Beyond the factors related to the stressor itself, such as intensity, duration or predictability, there are factors intrinsic to the individuals that are relevant to shape the stress response, such as age, sex and genetics. In this review, we examine the contribution of such intrinsic factors to the modulation of the stress response based on experimental rodent models of response to stress and discuss to what extent that knowledge can be potentially translated to humans. Effect of age in the stress response. Effect of sex in the stress response. Effect of genotype in the stress response.
Collapse
|
31
|
Bagot RC, Cates HM, Purushothaman I, Lorsch ZS, Walker DM, Wang J, Huang X, Schlüter OM, Maze I, Peña CJ, Heller EA, Issler O, Wang M, Song WM, Stein JL, Liu X, Doyle MA, Scobie KN, Sun HS, Neve RL, Geschwind D, Dong Y, Shen L, Zhang B, Nestler EJ. Circuit-wide Transcriptional Profiling Reveals Brain Region-Specific Gene Networks Regulating Depression Susceptibility. Neuron 2016; 90:969-83. [PMID: 27181059 DOI: 10.1016/j.neuron.2016.04.015] [Citation(s) in RCA: 237] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 03/16/2016] [Accepted: 04/11/2016] [Indexed: 12/30/2022]
Abstract
Depression is a complex, heterogeneous disorder and a leading contributor to the global burden of disease. Most previous research has focused on individual brain regions and genes contributing to depression. However, emerging evidence in humans and animal models suggests that dysregulated circuit function and gene expression across multiple brain regions drive depressive phenotypes. Here, we performed RNA sequencing on four brain regions from control animals and those susceptible or resilient to chronic social defeat stress at multiple time points. We employed an integrative network biology approach to identify transcriptional networks and key driver genes that regulate susceptibility to depressive-like symptoms. Further, we validated in vivo several key drivers and their associated transcriptional networks that regulate depression susceptibility and confirmed their functional significance at the levels of gene transcription, synaptic regulation, and behavior. Our study reveals novel transcriptional networks that control stress susceptibility and offers fundamentally new leads for antidepressant drug discovery.
Collapse
Affiliation(s)
- Rosemary C Bagot
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hannah M Cates
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Immanuel Purushothaman
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zachary S Lorsch
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Deena M Walker
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Junshi Wang
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Xiaojie Huang
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Oliver M Schlüter
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Ian Maze
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Catherine J Peña
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Elizabeth A Heller
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Orna Issler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Won-Min Song
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jason L Stein
- Department of Genetics and Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xiaochuan Liu
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Marie A Doyle
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kimberly N Scobie
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hao Sheng Sun
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rachael L Neve
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel Geschwind
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Li Shen
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Eric J Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
32
|
Malki K, Tosto MG, Pain O, Sluyter F, Mineur YS, Crusio WE, de Boer S, Sandnabba KN, Kesserwani J, Robinson E, Schalkwyk LC, Asherson P. Comparative mRNA analysis of behavioral and genetic mouse models of aggression. Am J Med Genet B Neuropsychiatr Genet 2016; 171B:427-36. [PMID: 26888158 DOI: 10.1002/ajmg.b.32424] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/22/2016] [Indexed: 11/06/2022]
Abstract
Mouse models of aggression have traditionally compared strains, most notably BALB/cJ and C57BL/6. However, these strains were not designed to study aggression despite differences in aggression-related traits and distinct reactivity to stress. This study evaluated expression of genes differentially regulated in a stress (behavioral) mouse model of aggression with those from a recent genetic mouse model aggression. The study used a discovery-replication design using two independent mRNA studies from mouse brain tissue. The discovery study identified strain (BALB/cJ and C57BL/6J) × stress (chronic mild stress or control) interactions. Probe sets differentially regulated in the discovery set were intersected with those uncovered in the replication study, which evaluated differences between high and low aggressive animals from three strains specifically bred to study aggression. Network analysis was conducted on overlapping genes uncovered across both studies. A significant overlap was found with the genetic mouse study sharing 1,916 probe sets with the stress model. Fifty-one probe sets were found to be strongly dysregulated across both studies mapping to 50 known genes. Network analysis revealed two plausible pathways including one centered on the UBC gene hub which encodes ubiquitin, a protein well-known for protein degradation, and another on P38 MAPK. Findings from this study support the stress model of aggression, which showed remarkable molecular overlap with a genetic model. The study uncovered a set of candidate genes including the Erg2 gene, which has previously been implicated in different psychopathologies. The gene networks uncovered points at a Redox pathway as potentially being implicated in aggressive related behaviors.
Collapse
Affiliation(s)
- Karim Malki
- King's College London, MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, London, United Kingdom
| | - Maria G Tosto
- King's College London, MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, London, United Kingdom.,Laboratory for Cognitive Investigations and Behavioral Genetics, Tomsk State University, Tomsk, Russia
| | - Oliver Pain
- Centre for Brain and Cognitive Development, Birkbeck, University of London, London, United Kingdom.,Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Frans Sluyter
- King's College London, MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, London, United Kingdom
| | - Yann S Mineur
- Department of Psychiatry, School of Medicine, Yale University, New Haven, Connecticut
| | - Wim E Crusio
- Aquitaine Institute for Cognitive and Integrative Neuroscience, University of Bordeaux, Bordeaux, France.,CNRS, Aquitaine Institute for Cognitive and Integrative Neuroscience, Bordeaux, France
| | - Sietse de Boer
- Groningen Institute for Evolutionary LifeSciences (GELIFES), University of Groningen, Groningen, The Netherlands
| | - Kenneth N Sandnabba
- Faculty of Arts, Psychology and Theology, Åbo Akademi University, Turku, Finland
| | - Jad Kesserwani
- King's College London, MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, London, United Kingdom
| | - Edward Robinson
- King's College London, MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, London, United Kingdom
| | - Leonard C Schalkwyk
- School of Biological Sciences, University of Essex, Colchester, United Kingdom
| | - Philip Asherson
- King's College London, MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, London, United Kingdom
| |
Collapse
|