1
|
Flynn LT, Gao WJ. DNA methylation and the opposing NMDAR dysfunction in schizophrenia and major depression disorders: a converging model for the therapeutic effects of psychedelic compounds in the treatment of psychiatric illness. Mol Psychiatry 2023; 28:4553-4567. [PMID: 37679470 PMCID: PMC11034997 DOI: 10.1038/s41380-023-02235-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/09/2023]
Abstract
Psychedelic compounds are being increasingly explored as a potential therapeutic option for treating several psychiatric conditions, despite relatively little being known about their mechanism of action. One such possible mechanism, DNA methylation, is a process of epigenetic regulation that changes gene expression via chemical modification of nitrogenous bases. DNA methylation has been implicated in the pathophysiology of several psychiatric conditions, including schizophrenia (SZ) and major depressive disorder (MDD). In this review, we propose alterations to DNA methylation as a converging model for the therapeutic effects of psychedelic compounds, highlighting the N-methyl D-aspartate receptor (NMDAR), a crucial mediator of synaptic plasticity with known dysfunction in both diseases, as an example and anchoring point. We review the established evidence relating aberrant DNA methylation to NMDAR dysfunction in SZ and MDD and provide a model asserting that psychedelic substances may act through an epigenetic mechanism to provide therapeutic effects in the context of these disorders.
Collapse
Affiliation(s)
- L Taylor Flynn
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
- MD/PhD program, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Wen-Jun Gao
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Prophylactic effect of Tongxieyaofang polysaccharide on depressive behavior in adolescent male mice with chronic unpredictable stress through the microbiome-gut-brain axis. Biomed Pharmacother 2023; 161:114525. [PMID: 36921537 DOI: 10.1016/j.biopha.2023.114525] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Major depression disorder is more common among adolescents and is a primary reason for suicide in adolescents. Some antidepressants are ineffective and may possess side effects. Therefore, developing an adolescent antidepressant is the need of the hour. We designed the stress model of adolescent male mice induced by chronic unpredictable stress (CUS). The mice were treated using Tongxieyaofang neutral polysaccharide (TXYF-NP), Tongxieyaofang acidic polysaccharide (TXYF-AP), TXYF-AP + TXYF-NP and fructooligosaccharide + galactooligosaccharides to determine their body weight, behavior, and serum hormone levels. RT-qPCR was used to detect the gene expression of Crhr1, Nr3c1, and Nr3c2 in the hypothalamus and hippocampus and the gene expression of glutamic acid and γ-aminobutyric acid-related receptors in the hippocampus. RT-qPCR, Western blot, and ELISA detected tryptophan metabolism in the colon, serum, and hippocampus. 16s rDNA helped sequence colon microflora, and non-targeted metabolomics enabled the collection of metabolic profiles of colon microflora. In adolescent male mice, CUS induced depression-like behavior, hypothalamic-pituitary-adrenal axis hyperactivity, hippocampal tissue damage, abnormal expression of its related receptors, and dysregulation of tryptophan metabolism. The 16s rDNA and non-targeted metabolomics revealed that CUS led to colon microflora disorder and bile acid metabolism abnormality. Tongxieyaofang polysaccharide could improve the bacterial community and bile acid metabolism disorder by upregulating the relative abundance of Lactobacillus gasseri, Lachnospiraceae bacterium 28-4, Bacteroides and Ruminococcaceae UCG-014 while preventing CUS-induced changes. TXYF-P can inhibit depression-like behavior due to CUS by regulating colonic microflora and restoring bile acid metabolism disorder. Thus, based on the different comparisons, TXYF-NP possessed the best effect.
Collapse
|
3
|
Cheng Z, Su J, Zhang K, Jiang H, Li B. Epigenetic Mechanism of Early Life Stress-Induced Depression: Focus on the Neurotransmitter Systems. Front Cell Dev Biol 2022; 10:929732. [PMID: 35865627 PMCID: PMC9294154 DOI: 10.3389/fcell.2022.929732] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Depression has an alarmingly high prevalence worldwide. A growing body of evidence indicates that environmental factors significantly affect the neural development and function of the central nervous system and then induce psychiatric disorders. Early life stress (ELS) affects brain development and has been identified as a major cause of depression. It could promote susceptibility to stress in adulthood. Recent studies have found that ELS induces epigenetic changes that subsequently affect transcriptional rates of differentially expressed genes. The epigenetic modifications involved in ELS include histone modifications, DNA methylation, and non-coding RNA. Understanding of these genetic modifications may identify mechanisms that may lead to new interventions for the treatment of depression. Many reports indicate that different types of ELS induce epigenetic modifications of genes involved in the neurotransmitter systems, such as the dopaminergic system, the serotonergic system, the gamma-aminobutyric acid (GABA)-ergic system, and the glutamatergic system, which further regulate gene expression and ultimately induce depression-like behaviors. In this article, we review the effects of epigenetic modifications on the neurotransmitter systems in depression-like outcomes produced by different types of ELS in recent years, aiming to provide new therapeutic targets for patients who suffer from depression.
Collapse
Affiliation(s)
- Ziqian Cheng
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
- Engineering Lab on Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
| | - Jingyun Su
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
- Engineering Lab on Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
| | - Kai Zhang
- Central Laboratory, The Second Hospital of Jilin University, Jilin University, Changchun, China
| | - Huiyi Jiang
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Huiyi Jiang, ; Bingjin Li,
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
- Engineering Lab on Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
- *Correspondence: Huiyi Jiang, ; Bingjin Li,
| |
Collapse
|
4
|
Waters RC, Gould E. Early Life Adversity and Neuropsychiatric Disease: Differential Outcomes and Translational Relevance of Rodent Models. Front Syst Neurosci 2022; 16:860847. [PMID: 35813268 PMCID: PMC9259886 DOI: 10.3389/fnsys.2022.860847] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 04/19/2022] [Indexed: 12/02/2022] Open
Abstract
It is now well-established that early life adversity (ELA) predisposes individuals to develop several neuropsychiatric conditions, including anxiety disorders, and major depressive disorder. However, ELA is a very broad term, encompassing multiple types of negative childhood experiences, including physical, sexual and emotional abuse, physical and emotional neglect, as well as trauma associated with chronic illness, family separation, natural disasters, accidents, and witnessing a violent crime. Emerging literature suggests that in humans, different types of adverse experiences are more or less likely to produce susceptibilities to certain conditions that involve affective dysfunction. To investigate the driving mechanisms underlying the connection between experience and subsequent disease, neuroscientists have developed several rodent models of ELA, including pain exposure, maternal deprivation, and limited resources. These studies have also shown that different types of ELA paradigms produce different but somewhat overlapping behavioral phenotypes. In this review, we first investigate the types of ELA that may be driving different neuropsychiatric outcomes and brain changes in humans. We next evaluate whether rodent models of ELA can provide translationally relevant information regarding links between specific types of experience and changes in neural circuits underlying dysfunction.
Collapse
Affiliation(s)
| | - Elizabeth Gould
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
| |
Collapse
|
5
|
Epigenome-wide association study of posttraumatic stress disorder identifies novel loci in U.S. military veterans. Transl Psychiatry 2022; 12:65. [PMID: 35177594 PMCID: PMC8854688 DOI: 10.1038/s41398-022-01822-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/26/2021] [Accepted: 01/14/2022] [Indexed: 01/23/2023] Open
Abstract
Posttraumatic stress disorder (PTSD) is a chronic and disabling psychiatric disorder prevalent in military veterans. Epigenetic mechanisms have been implicated in the etiology of PTSD, with DNA methylation being the most studied to identify novel molecular biomarkers associated with this disorder. We performed one of the largest single-sample epigenome-wide association studies (EWAS) of PTSD to date. Our sample included 1135 male European-American U.S. veterans who participated in the National Health and Resilience in Veterans Study (NHRVS). DNA was collected from saliva samples and the Illumina HumanMethylation EPIC BeadChip was used for the methylation analysis. PTSD was assessed using the PTSD Checklist. An EWAS was conducted using linear regression adjusted for age, cell-type proportions, first 10 principal components, and smoking status. After Bonferroni correction, we identified six genome-wide significant (GWS) CpG sites associated with past-month PTSD and three CpGs with lifetime PTSD (prange = 10-10-10-8). These CpG sites map to genes involved in immune function, transcription regulation, axonal guidance, cell signaling, and protein binding. Among these, SENP7, which is involved in transcription regulation and has been linked to risk-taking behavior and alcohol consumption in genome-wide association studies, replicated in an independent veteran cohort and was downregulated in medial orbitofrontal cortex of PTSD postmortem brain tissue. These findings suggest potential epigenetic biomarkers of PTSD that may help inform the pathophysiology of this disorder in veterans and other trauma-affected populations.
Collapse
|
6
|
Chen Y, Wang W, Fu X, Sun Y, Lv S, Liu L, Zhou P, Zhang K, Meng J, Zhang H, Zhang S. Investigation of the antidepressant mechanism of combined Radix Bupleuri and Radix Paeoniae Alba treatment using proteomics analysis of liver tissue. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1179:122858. [PMID: 34329891 DOI: 10.1016/j.jchromb.2021.122858] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/19/2021] [Accepted: 07/06/2021] [Indexed: 11/24/2022]
Abstract
Depression is a chronic, common mental illness characterized by depressed mood, anxiety, insomnia, cognitive impairment, and even suicidal tendency. In traditional Chinese medicine theory, the cause of depression is deemed to be "stagnation of liver qi". So relieving "stagnation of liver qi" is effective for depression. The combination of Radix Bupleuri and Radix Paeoniae Alba, which is used to soothe the liver and relieve depression, has antidepressant effects, but the mechanisms of the effects are still unclear. In this study, a rat model of chronic unpredictable mild stress was established as a model of depression, and proteomics analysis was used to explore the potential mechanisms of this combination in alleviating depression. Biological information analysis was performed on the selected differential proteins, and the enriched pathways mainly included the Jak-STAT signaling pathway, valine, leucine, and isoleucine degradation, and oxidative phosphorylation. The expression of key proteins included metallothionein-1, cyclin-dependent kinase, ubiquitin carboxyl-terminal hydrolase-1, and Cryab was further verified by western blotting, and the results which were consistent with the proteomics results, confirmed the reliability of the proteomic analysis. The antidepressant mechanism of combined Radix Bupleuri and Radix Paeoniae Alba treatment may be related to the oxidative stress response, neuroplasticity, the immune response, and neuroprotection.
Collapse
Affiliation(s)
- Yanyan Chen
- The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Wenran Wang
- The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Xin Fu
- Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Yonghui Sun
- Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Shaowa Lv
- Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Lei Liu
- Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Peng Zhou
- Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Ke Zhang
- Shenyang Pharmaceutical University, Shenyang 110000, China
| | - Jiannan Meng
- Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Hongcai Zhang
- Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China.
| | - Shuxiang Zhang
- Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China.
| |
Collapse
|
7
|
Bear T, Dalziel J, Coad J, Roy N, Butts C, Gopal P. The Microbiome-Gut-Brain Axis and Resilience to Developing Anxiety or Depression under Stress. Microorganisms 2021; 9:723. [PMID: 33807290 PMCID: PMC8065970 DOI: 10.3390/microorganisms9040723] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 03/29/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023] Open
Abstract
Episodes of depression and anxiety commonly follow the experience of stress, however not everyone who experiences stress develops a mood disorder. Individuals who are able to experience stress without a negative emotional effect are considered stress resilient. Stress-resilience (and its counterpart stress-susceptibility) are influenced by several psychological and biological factors, including the microbiome-gut-brain axis. Emerging research shows that the gut microbiota can influence mood, and that stress is an important variable in this relationship. Stress alters the gut microbiota and plausibly this could contribute to stress-related changes in mood. Most of the reported research has been conducted using animal models and demonstrates a relationship between gut microbiome and mood. The translational evidence from human clinical studies however is rather limited. In this review we examine the microbiome-gut-brain axis research in relation to stress resilience.
Collapse
Affiliation(s)
- Tracey Bear
- School of Food and Advanced Technology, Massey University, Palmerston North 4442, New Zealand;
- The New Zealand Institute for Plant and Food Research Limited, Palmerston North 4410, New Zealand; (C.B.); (P.G.)
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand; (J.D.); (N.R.)
| | - Julie Dalziel
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand; (J.D.); (N.R.)
- Smart Foods Innovation Centre of Excellence, AgResearch, Palmerston North 4442, New Zealand
| | - Jane Coad
- School of Food and Advanced Technology, Massey University, Palmerston North 4442, New Zealand;
| | - Nicole Roy
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand; (J.D.); (N.R.)
- Department of Human Nutrition, Otago University, Dunedin 9016, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1145, New Zealand
| | - Christine Butts
- The New Zealand Institute for Plant and Food Research Limited, Palmerston North 4410, New Zealand; (C.B.); (P.G.)
| | - Pramod Gopal
- The New Zealand Institute for Plant and Food Research Limited, Palmerston North 4410, New Zealand; (C.B.); (P.G.)
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand; (J.D.); (N.R.)
| |
Collapse
|
8
|
Analysis of Differentially Expressed Genes in the Dentate Gyrus and Anterior Cingulate Cortex in a Mouse Model of Depression. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5013565. [PMID: 33628784 PMCID: PMC7892236 DOI: 10.1155/2021/5013565] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/11/2020] [Accepted: 01/23/2021] [Indexed: 12/18/2022]
Abstract
Major depressive disorder (MDD) is a prevalent, chronic, and relapse-prone psychiatric disease. However, the intermediate molecules resulting from stress and neurological impairment in different brain regions are still unclear. To clarify the pathological changes in the dentate gyrus (DG) and anterior cingulate cortex (ACC) regions of the MDD brain, which are the most closely related to the disease, we investigated the published microarray profile dataset GSE84183 to identify unpredictable chronic mild stress- (UCMS-) induced differentially expressed genes (DEGs) in the DG and ACC regions. Based on the DEG data, functional annotation, protein-protein interaction, and transcription factor (TF) analyses were performed. In this study, 1071 DEGs (679 upregulated and 392 downregulated) and 410 DEGs (222 upregulated and 188 downregulated) were identified in DG and ACC, respectively. The pathways and GO terms enriched by the DEGs in the DG, such as cell adhesion, proteolysis, ion transport, transmembrane transport, chemical synaptic transmission, immune system processes, response to lipopolysaccharide, and nervous system development, may reveal the molecular mechanism of MDD. However, the DEGs in the ACC involved metabolic processes, proteolysis, visual learning, DNA methylation, innate immune responses, cell migration, and circadian rhythm. Sixteen hub genes in the DG (Fn1, Col1a1, Anxa1, Penk, Ptgs2, Cdh1, Timp1, Vim, Rpl30, Rps21, Dntt, Ptk2b, Jun, Avp, Slit1, and Sema5a) were identified. Eight hub genes in the ACC (Prkcg, Grin1, Syngap1, Rrp9, Grwd1, Pik3r1, Hnrnpc, and Prpf40a) were identified. In addition, eleven TFs (Chd2, Zmiz1, Myb, Etv4, Rela, Tcf4, Tcf12, Chd1, Mef2a, Ubtf, and Mxi1) were predicted to regulate more than two of these hub genes. The expression levels of ten randomly selected hub genes that were specifically differentially expressed in the MDD-like animal model were verified in the corresponding regions in the human brain. These hub genes and TFs may be regarded as potential targets for future MDD treatment strategies, thus aiding in the development of new therapeutic approaches to MDD.
Collapse
|
9
|
Ma M, Zhang X, Zhang Y, Su Y, Yan H, Tan H, Zhang D, Yue W. Childhood Maltreatment Was Correlated With the Decreased Cortical Function in Depressed Patients Under Social Stress in a Working Memory Task: A Pilot Study. Front Psychiatry 2021; 12:671574. [PMID: 34305677 PMCID: PMC8295536 DOI: 10.3389/fpsyt.2021.671574] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/25/2021] [Indexed: 01/30/2023] Open
Abstract
Background: Major depressive disorder (MDD) is a common psychiatric disorder associated with working memory (WM) impairment. Neuroimaging studies showed divergent results of the WM process in MDD patients. Stress could affect the occurrence and development of depression, in which childhood maltreatment played an important role. Methods: Thirty-seven MDD patients and 54 healthy control subjects were enrolled and completed a WM functional magnetic resonance imaging task with maintenance and manipulation conditions under stress and non-stress settings. We collected demographical and clinical data, using 17-item Hamilton Depression Scale (HAMD-17) and Childhood Trauma Questionnaire (CTQ) in MDD patients. In the WM task, we analyzed the main diagnosis effect and explored the correlation of impaired brain regions in MDD patients with CTQ and HAMD-17. Results: No group differences were found in the accuracy rate and reaction time between the two groups. MDD patients had lower brain activation in following regions (P FWE < 0.05). The left fusiform gyrus showed less activation in all conditions. The right supplementary motor area (SMA) exhibited decreased activation under non-stress. The anterior prefrontal cortex showed reduced activation during manipulation under stress, with the β estimations of the peak voxel showing significant group difference negatively correlated with childhood sex abuse (P Bonferroni < 0.05). Conclusions: In our pilot study, MDD patients had reduced brain activation, affecting emotional stimuli processing function, executive function, and cognitive control function. Childhood maltreatment might affect brain function in MDD. This work might provide some information for future studies on MDD.
Collapse
Affiliation(s)
- Mengying Ma
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, Beijing, China
| | - Xiao Zhang
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, Beijing, China
| | - Yuyanan Zhang
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, Beijing, China
| | - Yi Su
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, Beijing, China
| | - Hao Yan
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, Beijing, China
| | - Haoyang Tan
- Lieber Institute for Brain Development, Baltimore, MD, United States.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Dai Zhang
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, Beijing, China.,Lieber Institute for Brain Development, Baltimore, MD, United States
| | - Weihua Yue
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, Beijing, China.,PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| |
Collapse
|
10
|
Dygalo NN, Kalinina TS, Shishkina GT. Stress-induced expression pattern of glutamate signaling genes associated with anhedonia. Stress 2020; 23:700-707. [PMID: 32814471 DOI: 10.1080/10253890.2020.1812574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Chronic stress can predispose vulnerable individuals to mood disorders, including depression. Glutamate, one of the key participants in this process, may exert both pathological and therapeutic psycho-emotional effects. However, the role of expression of genes encoding proteins that provide glutamatergic signal is still unclear. In this study, we attempted to distinguish changes in expression of glutamatergic genes associated with stress-induced anhedonia, a core symptom of depression, from those related to other stress-related effects. For this, expression of genes was compared between rats after a short-term stress, which did not yet cause depressive-like symptoms, and animals exposed chronically to different stressors that produce anhedonia-like responses. The changes in gene expression induced by chronic restraint or forced swimming concomitantly with anhedonia development demonstrated similar for both stressors patterns. Main features of the expression patterns include the decrease in mRNA levels for AMPA and NMDA subunits in the midbrain and hippocampus that is consistent with the hypothesis that "monoamine (serotonin)-Glutamate/GABA long neural circuit" involved in mood regulation. The decrease in expression of these subunits in the midbrain may attenuate glutamatergic drive on the serotonergic neurons promoting a shift of excitation/inhibition balance between glutamate and GABA in the forebrain regions resulting in anhedonia. In general, changes in expression of multiple genes involved in glutamatergic neurotransmission in the forebrain and brainstem regions suggest that stress-induced anhedonia may result from the network dysfunction of this neurotransmitter system.
Collapse
Affiliation(s)
- Nikolay N Dygalo
- Laboratory of Functional Neurogenomics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Tatyana S Kalinina
- Laboratory of Functional Neurogenomics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, Novosibirsk, Russia
| | - Galina T Shishkina
- Laboratory of Functional Neurogenomics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, Novosibirsk, Russia
| |
Collapse
|
11
|
Cattaneo A, Cattane N, Scassellati C, D'Aprile I, Riva MA, Pariante CM. Convergent Functional Genomics approach to prioritize molecular targets of risk in early life stress-related psychiatric disorders. Brain Behav Immun Health 2020; 8:100120. [PMID: 34589878 PMCID: PMC8474593 DOI: 10.1016/j.bbih.2020.100120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/23/2020] [Accepted: 07/28/2020] [Indexed: 12/27/2022] Open
Abstract
There is an overwhelming evidence proving that mental disorders are not the product of a single risk factor - i.e. genetic variants or environmental factors, including exposure to maternal perinatal mental health problems or childhood adverse events - rather the product of a trajectory of cumulative and multifactorial insults occurring during development, such as exposures during the foetal life to adverse mental condition in the mother, or exposures to adverse traumatic events during childhood or adolescence. In this review, we aim to highlight the potential utility of a Convergent Functional Genomics (CFG) approach to clarify the complex brain-relevant molecular mechanisms and alterations induced by early life stress (ELS). We describe different studies based on CFG in psychiatry and neuroscience, and we show how this 'hypothesis-free' tool can prioritize a stringent number of genes modulated by ELS, that can be tested as potential candidates for Gene x Environment (GxE) interaction studies. We discuss the results obtained by using a CFG approach identifying FoxO1 as a gene where genetic variability can mediate the effect of an adverse environment on the development of depression. Moreover, we also demonstrate that FoxO1 has a functional relevance in stress-induced reduction of neurogenesis, and can be a potential target for the prevention or treatment of stress-related psychiatric disorders. Overall, we suggest that CFG approach could include trans-species and tissues data integration and we also propose the application of CFG to examine in depth and to prioritize top candidate genes that are affected by ELS across lifespan and generations.
Collapse
Affiliation(s)
- Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia
| | - Nadia Cattane
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia
| | - Catia Scassellati
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia
| | - Ilari D'Aprile
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Carmine Maria Pariante
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, United Kingdom
| |
Collapse
|
12
|
Hu X, Liu Y, Wu J, Liu Y, Liu W, Chen J, Yang F. Inhibition of P2X7R in the amygdala ameliorates symptoms of neuropathic pain after spared nerve injury in rats. Brain Behav Immun 2020; 88:507-514. [PMID: 32311494 DOI: 10.1016/j.bbi.2020.04.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/23/2020] [Accepted: 04/11/2020] [Indexed: 12/26/2022] Open
Abstract
The amygdala circuitry and P2X7 receptor (P2X7R) have both been shown to play important roles in the modulation of neuropathic pain (NP). However, little is known about the functional role of P2X7R in the amygdala for the regulation of NP. This study aims to evaluate the alleviative effect of intra-amygdala microinfusion of a pharmacological antagonist of P2X7R (A-438079) on NP and explore its possible mechanism of action. Male Sprague-Dawley rats were used to construct the animal model of NP through spared nerve injury (SNI). The SNI rats randomly received chronic bilateral microinjection of A-438079 (100 pmol/side) or saline into the amygdalae via cannulas. Mechanical paw withdrawal threshold (MWT) and thermal withdrawal duration (TWD) were measured by von Frey monofilaments. Besides, tail suspension test (TST), forced swimming test (FST), open field test (OFT) and sucrose preference test (SPT) were performed to assess depression- and anxiety-like behaviors. Immunofluorescence assay was employed to determine the levels of glial fibrillary acidic protein (GFAP), ionized calcium binding adaptor molecule 1 (IBA-1) and connexin 43 (Cx43) in the spinal cord. In addition, the change of growth associated protein 43 (GAP43) level in the spinal cord was assessed by Western blot. Our data showed that chronic treatment with A-438079 increased MWT and decreased TWD on days 11-21 post-SNI while decreased depression-like and anxiety-like behaviors. A-438079 administration significantly attenuated the elevated immunoreactivities of IBA-1 and GFAP in microglia and astrocytes after SNI. Furthermore, the decreased expression of GAP-43 in the spinal cord due to SNI was significantly attenuated by A-438079. However, when A-438079 and a pharmacological agonist (BzATP) of P2X7R were given simultaneously, all the effects caused by A-438079 alone were reversed. In brief, our study revealed the protective role of inhibiting P2X7R in the amygdala against symptoms associated with NP, possibly attributing to its inhibitory effects on spinal microglia and astrocytes.
Collapse
Affiliation(s)
- Xiaoling Hu
- Department of Anesthesiology, The First Affiliated Hospital of University of South China, Hunan Province 421001, China
| | - Yiming Liu
- Department of Anesthesiology, Affiliated Nanhua Hospital, University of South China, Hunan Province 421001, China
| | - Junting Wu
- Department of Anesthesiology, The First Affiliated Hospital of University of South China, Hunan Province 421001, China
| | - Yu Liu
- Department of Anesthesiology, The First Affiliated Hospital of University of South China, Hunan Province 421001, China
| | - Wenjie Liu
- Department of Anesthesiology, The First Affiliated Hospital of University of South China, Hunan Province 421001, China
| | - Ji Chen
- Department of Endocrinology, The First Affiliated Hospital of University of South China, Hunan Province 421001, China
| | - Fengrui Yang
- Department of Anesthesiology, The First Affiliated Hospital of University of South China, Hunan Province 421001, China; Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
13
|
Antontseva E, Bondar N, Reshetnikov V, Merkulova T. The Effects of Chronic Stress on Brain Myelination in Humans and in Various Rodent Models. Neuroscience 2020; 441:226-238. [DOI: 10.1016/j.neuroscience.2020.06.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 12/23/2022]
|
14
|
Adell A. Brain NMDA Receptors in Schizophrenia and Depression. Biomolecules 2020; 10:biom10060947. [PMID: 32585886 PMCID: PMC7355879 DOI: 10.3390/biom10060947] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 12/21/2022] Open
Abstract
N-methyl-D-aspartate (NMDA) receptor antagonists such as phencyclidine (PCP), dizocilpine (MK-801) and ketamine have long been considered a model of schizophrenia, both in animals and humans. However, ketamine has been recently approved for treatment-resistant depression, although with severe restrictions. Interestingly, the dosage in both conditions is similar, and positive symptoms of schizophrenia appear before antidepressant effects emerge. Here, we describe the temporal mechanisms implicated in schizophrenia-like and antidepressant-like effects of NMDA blockade in rats, and postulate that such effects may indicate that NMDA receptor antagonists induce similar mechanistic effects, and only the basal pre-drug state of the organism delimitates the overall outcome. Hence, blockade of NMDA receptors in depressive-like status can lead to amelioration or remission of symptoms, whereas healthy individuals develop psychotic symptoms and schizophrenia patients show an exacerbation of these symptoms after the administration of NMDA receptor antagonists.
Collapse
Affiliation(s)
- Albert Adell
- Institute of Biomedicine and Biotechnology of Cantabria, IBBTEC (CSIC-University of Cantabria), Calle Albert Einstein 22 (PCTCAN), 39011 Santander, Spain; or
- Biomedical Research Networking Center for Mental Health (CIBERSAM), 39011 Santander, Spain
| |
Collapse
|
15
|
de Araújo CM, Hudziak J, Crocetti D, Wymbs NF, Montalvo-Ortiz JL, Orr C, Albaugh MD, Althoff RR, O'Loughlin K, Holbrook H, Garavan H, Yang BZ, Mostofsky S, Jackowski A, Lee RS, Gelernter J, Kaufman J. Tubulin Polymerization Promoting Protein (TPPP) gene methylation and corpus callosum measures in maltreated children. Psychiatry Res Neuroimaging 2020; 298:111058. [PMID: 32120304 PMCID: PMC11079625 DOI: 10.1016/j.pscychresns.2020.111058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/16/2020] [Accepted: 02/19/2020] [Indexed: 12/21/2022]
Abstract
The goal of the current study was to evaluate the impact of Tubulin Polymerization Promoting Protein (TPPP) methylation on structural and fractional anisotropy (FA) corpus callosum (CC) measures. TPPP is involved in the development of white matter tracts in the brain and was implicated in stress-related psychiatric disorders in an unbiased whole epigenome methylation study. The cohort included 63 participants (11.73 y/o ±1.91) from a larger study investigating risk and resilience in maltreated children. Voxel-based morphometry (VBM) was used to process the structural data, fractional anisotropy (FA) was determined using an atlas-based approach, and DNA specimens were derived from saliva in two batches using the 450 K (N = 39) and 850 K (N = 24) Illumina arrays, with the data from each batch analyzed separately. After controlling for multiple comparisons and relevant covariates (e.g., demographics, brain volume, cell composition, 3 PCs), 850 K derived TPPP methylation values, in interaction with a dimensional measure of children's trauma experiences, predicted left and right CC body volumes and genu, body and splenium FA (p < .007, all comparisons). The findings in the splenium replicated in subjects with the 450 K data. The results extend prior investigations and suggest a role for TPPP in brain changes associated with stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Célia Maria de Araújo
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, United States; Universidade Federal de São Paulo, São Paulo, Brazil
| | - James Hudziak
- Vermont Center for Children, Youth, and Families, Department of Psychiatry, University of Vermont, Burlington, VT, United States
| | - Deana Crocetti
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, United States
| | - Nicholas F Wymbs
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, United States
| | | | - Catherine Orr
- Vermont Center for Children, Youth, and Families, Department of Psychiatry, University of Vermont, Burlington, VT, United States
| | - Matthew D Albaugh
- Vermont Center for Children, Youth, and Families, Department of Psychiatry, University of Vermont, Burlington, VT, United States
| | - Robert R Althoff
- Vermont Center for Children, Youth, and Families, Department of Psychiatry, University of Vermont, Burlington, VT, United States
| | - Kerry O'Loughlin
- Vermont Center for Children, Youth, and Families, Department of Psychiatry, University of Vermont, Burlington, VT, United States
| | - Hannah Holbrook
- Vermont Center for Children, Youth, and Families, Department of Psychiatry, University of Vermont, Burlington, VT, United States
| | - Hugh Garavan
- Vermont Center for Children, Youth, and Families, Department of Psychiatry, University of Vermont, Burlington, VT, United States
| | - Bao-Zhu Yang
- Department of Psychiatry, Yale University, New Haven, CT, United States
| | - Stewart Mostofsky
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, United States; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, , United States; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | | | - Richard S Lee
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Joel Gelernter
- Department of Psychiatry, Yale University, New Haven, CT, United States; Veterans Administration, West Haven, CT, United States
| | - Joan Kaufman
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States; Center for Child and Family Traumatic Stress, Kennedy Krieger Institute, Baltimore, MD, United States.
| |
Collapse
|
16
|
Meng J, Wang DM, Luo LL. CTRP3 acts as a novel regulator in depressive-like behavior associated inflammation and apoptosis by meditating p38 and JNK MAPK signaling. Biomed Pharmacother 2019; 120:109489. [PMID: 31629950 DOI: 10.1016/j.biopha.2019.109489] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/18/2019] [Accepted: 09/22/2019] [Indexed: 10/25/2022] Open
Abstract
Depression is a complicated etiological pattern, and its pathology and effective treatments are highly limited.C1q-tumor necrosis factor-related protein-3 (CTRP3) is an adipokine, playing crucial roles in metabolic regulatory properties. However, the effects of CTRP3 on depression are largely unknown. In the present study, we found that CTRP3 expression levels were markedly reduced in hippocampus of mice with depression induced by chronic unpredictable mild stress (CUMS). In mouse model with depression, CTRP3-deficient mice aggravated depression-associated behaviors, as evidenced by the reduced locomotor activity and sucrose consumption, while the elevated immobility time in the tail suspension test (TST) and forced swimming test (FST). Moreover, CUMS-induced neuron death and increased expression of cleaved Caspase-3 were significantly accelerated by CTRP3 knockout. Furthermore, CTRP3 deletion intensified pro-inflammatory response in CUMS-exposed mice, which was associated with the activation of nuclear factor-κB(NF-κB) signaling. The activity of mitogen-activated protein kinases (MAPKs), including p38 and JNK, was further promoted in hippocampus of CTRP3-knockout mice with CUMS exposure. In contrast,CTRP3 over-expression showed anti-apoptotic and anti-inflammatory effects in lipopolysaccharide (LPS)-treated microglial cells. Importantly, the in vitro experiments demonstrated that CTRP3 knockdown-exacerbated apoptosis and inflammatory responsewere remarkably abrogated by the blockage of p38 and JNK signaling pathways in microglia stimulated by LPS. Next, in CUMS-exposed mice with CTRP3 deficiency, suppressing p38 and JNK markedly alleviated depressive-like behavior,hippocampal neuron death, apoptosis and inflammation. Therefore, CTRP3 may be an innovative therapeutic target for treating patients with depression through regulating p38 and JNK signaling.
Collapse
Affiliation(s)
- Jing Meng
- Department of Geriatrics, Wuhan Mental Health Center, Wuhan, 430022, China
| | - Dong-Ming Wang
- Department of Geriatric Psychiatry, Qingdao Mental Heath Center, Qingdao, 266034, China
| | - Li-Ling Luo
- Department of Psychosomatic, The Fourth People's Hospital of Shaanxi, Xi'an, 710043, China.
| |
Collapse
|
17
|
Music exposure attenuates anxiety- and depression-like behaviors and increases hippocampal spine density in male rats. Behav Brain Res 2019; 372:112023. [DOI: 10.1016/j.bbr.2019.112023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/18/2019] [Accepted: 06/07/2019] [Indexed: 01/14/2023]
|
18
|
Corrêa T, Feltes BC, Riegel M. Integrated analysis of the critical region 5p15.3-p15.2 associated with cri-du-chat syndrome. Genet Mol Biol 2019; 42:186-196. [PMID: 30985858 PMCID: PMC6687350 DOI: 10.1590/1678-4685-gmb-2018-0173] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 07/29/2018] [Indexed: 11/21/2022] Open
Abstract
Cri-du-chat syndrome (CdCs) is one of the most common contiguous gene syndromes, with an incidence of 1:15,000 to 1:50,000 live births. To better understand the etiology of CdCs at the molecular level, we investigated theprotein-protein interaction (PPI) network within the critical chromosomal region 5p15.3-p15.2 associated with CdCs using systemsbiology. Data were extracted from cytogenomic findings from patients with CdCs. Based on clinical findings, molecular characterization of chromosomal rearrangements, and systems biology data, we explored possible genotype-phenotype correlations involving biological processes connected with CdCs candidate genes. We identified biological processes involving genes previously found to be associated with CdCs, such as TERT, SLC6A3, and CTDNND2, as well as novel candidate proteins with potential contributions to CdCs phenotypes, including CCT5, TPPP, MED10, ADCY2, MTRR, CEP72, NDUFS6, and MRPL36. Although further functional analyses of these proteins are required, we identified candidate proteins for the development of new multi-target genetic editing tools to study CdCs. Further research may confirm those that are directly involved in the development of CdCs phenotypes and improve our understanding of CdCs-associated molecular mechanisms.
Collapse
Affiliation(s)
- Thiago Corrêa
- Post-Graduate Program in Genetics and Molecular Biology,
Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bruno César Feltes
- Institute of Informatics, Universidade Federal do Rio Grande
do Sul, Porto Alegre, RS, Brazil
| | - Mariluce Riegel
- Post-Graduate Program in Genetics and Molecular Biology,
Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Medical Genetics Service, Hospital de Clínicas de Porto
Alegre, Porto Alegre, RS, Brazil
| |
Collapse
|
19
|
Li MM, Zhou P, Chen XD, Xu HS, Wang J, Chen L, Zhang N, Liu N. NO in the dPAG modulates panic-like responses and ASIC1a expression in the prefrontal cortex and hippocampus in mice. Biochem Biophys Res Commun 2019; 511:274-279. [PMID: 30770101 DOI: 10.1016/j.bbrc.2019.02.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 02/04/2019] [Indexed: 01/20/2023]
Abstract
Panic disorder (PD) is a multifactorial neuropsychiatric disorder. Our previous study has demonstrated that the nitric oxide (NO) pathway and the acid-sensing ion channel 1a (ASIC1a) level in the dorsal midbrain periaqueductal gray (dPAG) are involved in the modulation of panic-like responses. In addition, the prefrontal cortex (PFC) and the hippocampus also play a role in panic-like responses. However, no studies have investigated the protein level of ASIC1a in the PFC and hippocampus in a mouse model of panic-like disorders after alteration of the NO pathway in the dPAG. We investigated the production of a panic attack with intra-dPAG injections of SNAP, an NO donor, and 7-NI, an nNOS inhibitor. Moreover, we measured ASIC1a protein levels in the PFC and hippocampus. The rat exposure test (RET) is frequently used as an animal model of panic. In our study, C57BL/6 mice received an intra-dPAG injection of SNAP or 7-NI before RET; neurobehavioral tests were then conducted, followed by mechanistic evaluation through western blot analysis in the PFC and hippocampus. An intra-dPAG infusion of SNAP significantly increased the panic-like effect, whereas treatment with 7-NI decreased fear behavior. Mice treated with SNAP/7-NI showed significantly increased/decreased ASIC1a expression in the PFC, and a decreasing/increasing trend in the hippocampus. The present study suggests that the NO pathway in the dPAG plays a key role in panic-like responses in mice confronted by a rat, further, NO intra-dPAG injection also modulates the ASIC1a expression levels in the PFC and hippocampus.
Collapse
Affiliation(s)
- Meng-Meng Li
- Medical School, Nanjing University, Nanjing, 210093, China
| | - Ping Zhou
- Department of Medical Psychology, Nanjing Medical University, Affiliated Nanjing Brain Hospital, Nanjing, 210029, China
| | - Xiao-Dong Chen
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, 210029, China
| | - Huai-Sha Xu
- Medical School, Nanjing University, Nanjing, 210093, China
| | - Jun Wang
- Department of Toxicology, The Key Lab of Modern Toxicology (NJMU), Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Ling Chen
- State Key Laboratory of Reproductive Medicine, Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
| | - Ning Zhang
- Department of Medical Psychology, Nanjing Medical University, Affiliated Nanjing Brain Hospital, Nanjing, 210029, China
| | - Na Liu
- Department of Medical Psychology, Nanjing Medical University, Affiliated Nanjing Brain Hospital, Nanjing, 210029, China.
| |
Collapse
|
20
|
Ma W, Tang C, Hu H, Zhang F, Wang X, Wu X, Zhang W, Wang X, Ma H, Li Z, Dong Y, Yang Z, Feng S, Tian L, Gao Y. Advance in Tissue Differentiation and its Regulatory Mechanisms by Master Proteins of Nervous System during Weaning. Curr Protein Pept Sci 2019; 20:683-689. [PMID: 30678621 DOI: 10.2174/1389203720666190125101039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 12/30/2018] [Accepted: 01/13/2019] [Indexed: 11/22/2022]
Abstract
Weaning is a critical period for the growth and development of mammals, in which various physiological and biochemical indicators of the body have undergone great changes. The development, differentiation, and maturation of the nervous system are regulated by many proteins. Changes in related proteins affect the physiological functions of the nervous system. However, the regulation of selfrenewal and differentiation of the nervous system at this stage is still poorly understood. The mechanism of differentiation and regulation of the major proteins in the nervous system during this special period of weaning remains to be investigated. Therefore, this paper aims to summarize the alteration of the nervous system during weaning and provide the basis for subsequent research.
Collapse
Affiliation(s)
- Wenyu Ma
- College of PIWEI institute, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.,College of Pharmacy, Shihezi University, Shihezi, 832001, China
| | - Chengfang Tang
- College of Pharmacy, Shihezi University, Shihezi, 832001, China
| | - Huiling Hu
- College of PIWEI institute, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Fenglian Zhang
- Department of Operating Theatre, Binzhou People's Hospital, Binzhou, 256610, China
| | - Xuanying Wang
- Department of Operating Theatre, Binzhou People's Hospital, Binzhou, 256610, China
| | - Xiaoting Wu
- Department of Operating Theatre, Binzhou People's Hospital, Binzhou, 256610, China
| | - Wenjian Zhang
- Department of Operating Theatre, Binzhou People's Hospital, Binzhou, 256610, China
| | - Xiaoxia Wang
- Department of Operating Theatre, Binzhou People's Hospital, Binzhou, 256610, China
| | - Huazhi Ma
- Department of Rheumatology, Binzhou People's Hospital, Binzhou, 256610, China
| | - Zhihao Li
- College of PIWEI institute, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yanbin Dong
- College of PIWEI institute, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zehong Yang
- College of PIWEI institute, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Shixiu Feng
- Key Laboratory of Southern Subtropical Plant Diversity, Shenzhen Fairy Lake Botanical Garden, Chinese Academy of Sciences, Shenzhen, 518004, China
| | - Liping Tian
- College of Pharmacy, Shihezi University, Shihezi, 832001, China
| | - Yong Gao
- College of PIWEI institute, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| |
Collapse
|
21
|
Avecilla V, Avecilla A. Inhibitor of DNA-Binding/Differentiation Proteins and Environmental Toxicants: Genomic Impact on the Onset of Depressive Dysfunction. ACTA ACUST UNITED AC 2019; 7:medsci7010007. [PMID: 30634536 PMCID: PMC6358799 DOI: 10.3390/medsci7010007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/18/2018] [Accepted: 01/07/2019] [Indexed: 12/21/2022]
Abstract
The ongoing growth of the international occurrence of depression and its ability to co-occur with other serious medical disorders, such as heart disease, cancer, diabetes, and Parkinson’s disease, is a current public health problem. Inhibitor of DNA-Binding/Differentiation (ID) proteins are part of a group of transcriptional factors that have shown involvement in neurocognitive disorders and, therefore, may have influence on depressive disorders. Previously, it has been established that exposure to environmental estrogenic endocrine disruptors (EEDs), such as polychlorinated biphenyls (PCBs) and bisphenol A (BPA), have played an important role in the modulation of depressive disorders. Hence, based on many studies, we consider the impact of these environmental pollutants on the group of ID proteins and how they impact depressive outcomes. Improved knowledge of how ID proteins interact with depressive disorders, through EED exposure, will contribute essential evidence that can further benefit our public health community with innovative knowledge to prevent these types of mental illnesses.
Collapse
Affiliation(s)
- Vincent Avecilla
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, USA.
- Celgene Corporation, Summit, NJ 07901, USA.
| | - Andrea Avecilla
- Department of Clinical Psychology, University of Massachusetts Dartmouth, North Dartmouth, MA 02747, USA.
| |
Collapse
|
22
|
Contribution of Inhibitor of Differentiation and Estrogenic Endocrine Disruptors to Neurocognitive Disorders. Med Sci (Basel) 2018; 6:medsci6030061. [PMID: 30081481 PMCID: PMC6165108 DOI: 10.3390/medsci6030061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 01/17/2023] Open
Abstract
The devastating growth in the worldwide frequency of neurocognitive disorders and its allied difficulties, such as decline in memory, spatial competency, and ability to focus, poses a significant psychological public health problem. Inhibitor of differentiation (ID) proteins are members of a family of helix-loop-helix (HLH) transcription factors. ID proteins have been demonstrated to be involved in neurodevelopmental and depressive diseases and, thus, may influence neurocognitive deficiencies due to environmental exposure. Previously, it has been demonstrated that environmental factors, such as estrogenic endocrine disruptors (EEDs), have played an essential role in the influence of various neurocognitive disorders such as Alzheimer’s, dementia, and Parkinson’s disease. Based on this increasing number of reports, we consider the impact of these environmental pollutants on ID proteins. Better understanding of how these ID proteins by which EED exposure can affect neurocognitive disorders in populations will prospectively deliver valuable information in the impediment and regulation of these diseases linked with environmental factor exposure.
Collapse
|
23
|
Alavian-Ghavanini A, Lin PI, Lind PM, Risén Rimfors S, Halin Lejonklou M, Dunder L, Tang M, Lindh C, Bornehag CG, Rüegg J. Prenatal Bisphenol A Exposure is Linked to Epigenetic Changes in Glutamate Receptor Subunit Gene Grin2b in Female Rats and Humans. Sci Rep 2018; 8:11315. [PMID: 30054528 PMCID: PMC6063959 DOI: 10.1038/s41598-018-29732-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 07/17/2018] [Indexed: 12/15/2022] Open
Abstract
Bisphenol A (BPA) exposure has been linked to neurodevelopmental disorders and to effects on epigenetic regulation, such as DNA methylation, at genes involved in brain function. High doses of BPA have been shown to change expression and regulation of one such gene, Grin2b, in mice. Yet, if such changes occur at relevant doses in animals and humans has not been addressed. We investigated if low-dose developmental BPA exposure affects DNA methylation and expression of Grin2b in brains of adult rats. Furthermore, we assessed associations between prenatal BPA exposure and Grin2b methylation in 7-year old children. We found that Grin2b mRNA expression was increased and DNA methylation decreased in female, but not in male rats. In humans, prenatal BPA exposure was associated with increased methylation levels in girls. Additionally, low APGAR scores, a predictor for increased risk for neurodevelopmental diseases, were associated with higher Grin2b methylation levels in girls. Thus, we could link developmental BPA exposure and low APGAR scores to changes in the epigenetic regulation of Grin2b, a gene important for neuronal function, in a sexual dimorphic fashion. Discrepancies in exact locations and directions of the DNA methylation change might reflect differences between species, analysed tissues, exposure level and/or timing.
Collapse
Affiliation(s)
- Ali Alavian-Ghavanini
- Swetox, Karolinska Institutet, Unit of Toxicology Sciences, Forskargatan 20, 151 36, Södertälje, Sweden
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Molecular Medicine (CMM), 171 64, Solna, Sweden
| | - Ping-I Lin
- Karlstad University, Department of Health Sciences, 651 88, Karlstad, Sweden
| | - P Monica Lind
- Uppsala University, Department of Medical Sciences, Occupational and Environmental Medicine, 751 85, Uppsala, Sweden
| | - Sabina Risén Rimfors
- Swetox, Karolinska Institutet, Unit of Toxicology Sciences, Forskargatan 20, 151 36, Södertälje, Sweden
| | - Margareta Halin Lejonklou
- Uppsala University, Department of Medical Sciences, Occupational and Environmental Medicine, 751 85, Uppsala, Sweden
| | - Linda Dunder
- Uppsala University, Department of Medical Sciences, Occupational and Environmental Medicine, 751 85, Uppsala, Sweden
| | - Mandy Tang
- Swetox, Karolinska Institutet, Unit of Toxicology Sciences, Forskargatan 20, 151 36, Södertälje, Sweden
| | - Christian Lindh
- Lund University, Division of Occupational and Environmental Medicine, Lund University, 221 85, Lund, Sweden
| | - Carl-Gustaf Bornehag
- Karlstad University, Department of Health Sciences, 651 88, Karlstad, Sweden
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joëlle Rüegg
- Swetox, Karolinska Institutet, Unit of Toxicology Sciences, Forskargatan 20, 151 36, Södertälje, Sweden.
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Molecular Medicine (CMM), 171 64, Solna, Sweden.
| |
Collapse
|
24
|
Behavioral Changes in Mice Lacking Interleukin-33. eNeuro 2017; 4:eN-NWR-0147-17. [PMID: 29379874 PMCID: PMC5788055 DOI: 10.1523/eneuro.0147-17.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 11/21/2017] [Accepted: 12/03/2017] [Indexed: 12/26/2022] Open
Abstract
Interleukin (IL)-33 is a member of the IL-1 family of cytokines. IL-33 is expressed in nuclei and secreted as alarmin upon cellular damage to deliver a danger signal to the surrounding cells. Previous studies showed that IL-33 is expressed in the brain and that it is involved in neuroinflammatory and neurodegenerative processes in both humans and rodents. Nevertheless, the role of IL-33 in physiological brain function and behavior remains unclear. Here, we have investigated the behaviors of mice lacking IL-33 (Il33−/− mice). IL-33 is constitutively expressed throughout the adult mouse brain, mainly in oligodendrocyte-lineage cells and astrocytes. Notably, Il33−/− mice exhibited reduced anxiety-like behaviors in the elevated plus maze (EPM) and the open field test (OFT), as well as deficits in social novelty recognition, despite their intact sociability, in the three-chamber social interaction test. The immunoreactivity of c-Fos proteins, an indicator of neuronal activity, was altered in several brain regions implicated in anxiety-related behaviors, such as the medial prefrontal cortex (mPFC), amygdala, and piriform cortex (PCX), in Il33−/− mice after the EPM. Altered c-Fos immunoreactivity in Il33−/− mice was not correlated with IL-33 expression in wild-type (WT) mice nor was IL-33 expression affected by the EPM in WT mice. Thus, our study has revealed that Il33−/− mice exhibit multiple behavioral deficits, such as reduced anxiety and impaired social recognition. Our findings also indicate that IL-33 may regulate the development and/or maturation of neuronal circuits, rather than control neuronal activities in adult brains.
Collapse
|
25
|
Carlyle BC, Kitchen RR, Kanyo JE, Voss EZ, Pletikos M, Sousa AMM, Lam TT, Gerstein MB, Sestan N, Nairn AC. A multiregional proteomic survey of the postnatal human brain. Nat Neurosci 2017; 20:1787-1795. [PMID: 29184206 PMCID: PMC5894337 DOI: 10.1038/s41593-017-0011-2] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 09/27/2017] [Indexed: 12/13/2022]
Abstract
Detailed observations of transcriptional, translational and post-translational events in the human brain are essential to improving our understanding of its development, function and vulnerability to disease. Here, we exploited label-free quantitative tandem mass-spectrometry to create an in-depth proteomic survey of regions of the postnatal human brain, ranging in age from early infancy to adulthood. Integration of protein data with existing matched whole-transcriptome sequencing (RNA-seq) from the BrainSpan project revealed varied patterns of protein-RNA relationships, with generally increased magnitudes of protein abundance differences between brain regions compared to RNA. Many of the differences amplified in protein data were reflective of cytoarchitectural and functional variation between brain regions. Comparing structurally similar cortical regions revealed significant differences in the abundances of receptor-associated and resident plasma membrane proteins that were not readily observed in the RNA expression data.
Collapse
Affiliation(s)
- Becky C Carlyle
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Robert R Kitchen
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Molecular Biophysics & Biochemistry, Yale School of Medicine, New Haven, CT, USA
| | - Jean E Kanyo
- W.M. Keck Biotechnology Resource Laboratory, Yale School of Medicine, New Haven, CT, USA
| | - Edward Z Voss
- W.M. Keck Biotechnology Resource Laboratory, Yale School of Medicine, New Haven, CT, USA
| | - Mihovil Pletikos
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - André M M Sousa
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - TuKiet T Lam
- Department of Molecular Biophysics & Biochemistry, Yale School of Medicine, New Haven, CT, USA
- W.M. Keck Biotechnology Resource Laboratory, Yale School of Medicine, New Haven, CT, USA
| | - Mark B Gerstein
- Department of Molecular Biophysics & Biochemistry, Yale School of Medicine, New Haven, CT, USA
| | - Nenad Sestan
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA.
- Departments of Genetics and Psychiatry, Section of Comparative Medicine, and Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA.
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT, USA.
| | - Angus C Nairn
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
26
|
Contribution of Inhibitor of DNA Binding/Differentiation-3 and Endocrine Disrupting Chemicals to Pathophysiological Aspects of Chronic Disease. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6307109. [PMID: 28785583 PMCID: PMC5530454 DOI: 10.1155/2017/6307109] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/15/2017] [Accepted: 05/29/2017] [Indexed: 12/12/2022]
Abstract
The overwhelming increase in the global incidence of obesity and its associated complications such as insulin resistance, atherosclerosis, pulmonary disease, and degenerative disorders including dementia constitutes a serious public health problem. The Inhibitor of DNA Binding/Differentiation-3 (ID3), a member of the ID family of transcriptional regulators, has been shown to play a role in adipogenesis and therefore ID3 may influence obesity and metabolic health in response to environmental factors. This review will highlight the current understanding of how ID3 may contribute to complex chronic diseases via metabolic perturbations. Based on the increasing number of reports that suggest chronic exposure to and accumulation of endocrine disrupting chemicals (EDCs) within the human body are associated with metabolic disorders, we will also consider the impact of these chemicals on ID3. Improved understanding of the ID3 pathways by which exposure to EDCs can potentiate complex chronic diseases in populations with metabolic disorders (obesity, metabolic syndrome, and glucose intolerance) will likely provide useful knowledge in the prevention and control of complex chronic diseases associated with exposure to environmental pollutants.
Collapse
|