1
|
Arvind A, Sreelekshmi S, Dubey N. Genetic, Epigenetic, and Hormonal Regulation of Stress Phenotypes in Major Depressive Disorder: From Maladaptation to Resilience. Cell Mol Neurobiol 2025; 45:29. [PMID: 40138049 PMCID: PMC11947386 DOI: 10.1007/s10571-025-01549-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
Major Depressive Disorder (MDD) is a complex psychiatric disorder with varied molecular mechanisms underlying its aetiology, diagnosis, and treatment. This review explores the crucial roles of stress, genetics, epigenetics, and hormones in shaping susceptibility and resilience to mood disorders. We discuss how acute stress can be beneficial, while prolonged stress disrupts brain function, leading to MDD. The review also highlights the significance of various animal models in understanding depression pathophysiology, including zebrafish, mice, and rats, which exhibit distinct sex differences in stress responses. Furthermore, we delve into the molecular bases of susceptible and resilient phenotypes, focusing on genetic aspects such as gene polymorphisms, mutations, and telomere length alterations. The review also examines epigenetic aspects including DNA methylation, histone acetylation and deacetylation, histone methylation and HMTs, and miRNA, which contribute to the development of MDD. Additionally, we explore the role of hormones such as estrogen, progesterone, and prolactin in modulating stress responses and influencing MDD susceptibility and resilience. Finally, we discuss the clinical implications of these findings, including recent clinical methods for determining MDD susceptibility and resiliency phenotypes. By consolidating the current knowledge and insights, this review aims to provide a comprehensive understanding of the molecular basis of susceptibility and resilience in mood disorders, contributing to the ongoing efforts in combating this debilitating disorder.
Collapse
Affiliation(s)
- Anushka Arvind
- Dr Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, 500046, Telangana, India
| | - S Sreelekshmi
- Endocrinology Unit, Department of Zoology, Madras Christian College, East Tambaram, Chennai, 600059, Tamil Nadu, India
| | - Neelima Dubey
- Dr Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, 500046, Telangana, India.
| |
Collapse
|
2
|
Gyles T, Parise EM, Estill MS, Browne CJ, Shen L, Nestler EJ, Torres-Berrío A. Transcriptional Profiles in Nucleus Accumbens of Antidepressant Resistance in Chronically Stressed Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.17.643727. [PMID: 40166343 PMCID: PMC11956914 DOI: 10.1101/2025.03.17.643727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Unsuccessful response to several courses of antidepressants is a core feature of treatment-resistant depression (TRD), a severe condition that affects a third of patients with depression treated with conventional pharmacotherapy. However, the molecular mechanisms underlying TRD remain poorly understood. Here, we assessed the successful vs. unsuccessful response to ketamine (KET) in chronically stressed mice that failed to respond to initial treatment with fluoxetine (FLX) as a rodent model of TRD and characterized the associated transcriptional profiles in the nucleus accumbens (NAc) using RNA-sequencing. We observed that failed treatment with FLX exerts a priming effect that promotes behavioral and transcriptional responses to subsequent ketamine treatment. We also identified specific gene networks that are linked to both susceptibility to stress and resistance to antidepressant response. Collectively, these findings offer valuable insights into the molecular mechanisms underlying antidepressant resistance and help address a critical gap in preclinical models of TRD.
Collapse
|
3
|
Zhu Y, Tong J, Jiang J, Shi K, Xie J, Zhu Y, Li Y, Xu Y. Suppression of miR-218 promotes SOCS3 expression to alleviate cognitive impairment and white matter injury after chronic cerebral hypoperfusion. Metab Brain Dis 2025; 40:147. [PMID: 40080253 DOI: 10.1007/s11011-025-01572-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
Chronic cerebral hypoperfusion-induced white matter injury (WMI) is a significant cause of vascular cognitive impairment. Emerging evidence suggests that miR-218 may be involved in the pathogenesis of WMI. However, understanding of the relationship between miR-218 and chronic hypoperfusion-induced WMI remains insufficient. Our study investigated the relationship between miR-218 and chronic hypoperfusion-induced WMI at clinical, animal, and cellular levels. We found that serum miR-218 expression was elevated in clinical WMI patients, had a certain diagnostic efficacy for WMI, and was correlated with the degree of WMI, cognitive scores, and serum inflammatory factors. In addition, we constructed a mouse model with bilateral carotid artery stenosis (BCAS) to simulate chronic hypoperfusion-induced WMI and detected an increase in miR-218 expression in the white matter of BCAS mice. Following administration of Lv-sh-miR-218 to the white matter of BCAS mice, improvements were observed in both cognitive impairment and WMI. Furthermore, Lv-sh-miR-218 also reduced M1 polarization of microglia and neuroinflammation within the white matter. Subsequently, we confirmed that SOCS3 is the specific target of miR-218 through bioinformatics analysis and luciferase reporter gene assays. Injection of LV-SOCS3 into the white matter also led to improvements in cognitive impairment and WMI in BCAS mice, along with reduced M1 polarization of microglia and neuroinflammation. Moreover, in primary cultured microglia cells, we demonstrated that after chronic hypoxia, miR-218 regulates inflammatory factors through the SOCS3/STAT3 pathway. In summary, our current results indicate a strong correlation between elevated miR-218 levels and chronic hypoperfusion-induced WMI, and downregulation of miR-218 expression can improve neuroinflammation by upregulating SOCS3, thereby ameliorating WMI and cognitive impairment.
Collapse
Affiliation(s)
- Yuanyuan Zhu
- Department of Geriatrics, The Affiliated Yixing Hospital of Jiangsu University, Yixing, 214221, Jiangsu, China
- Department of Neuroimaging Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Juan Tong
- Department of Neurology, Dianjiang People's Hospital of Chongqing, Chongqing, Chongqing, 408300, China
| | - Jianzhong Jiang
- Department of Geriatrics, The Affiliated Yixing Hospital of Jiangsu University, Yixing, 214221, Jiangsu, China
| | - Keyun Shi
- Department of Geriatrics, The Affiliated Yixing Hospital of Jiangsu University, Yixing, 214221, Jiangsu, China
| | - Jing Xie
- Department of Geriatrics, The Affiliated Yixing Hospital of Jiangsu University, Yixing, 214221, Jiangsu, China
| | - Yan Zhu
- Department of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
- Department of Neuroimaging Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yuefeng Li
- Department of Radiology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, 214221, Jiangsu, China.
- Department of Neuroimaging Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| | - Yuhao Xu
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China.
- Department of Neuroimaging Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
4
|
Rodrigues B, Leitão RA, Santos M, Trofimov A, Silva M, Inácio ÂS, Abreu M, Nobre RJ, Costa J, Cardoso AL, Milosevic I, Peça J, Oliveiros B, Pereira de Almeida L, Pinheiro PS, Carvalho AL. MiR-186-5p inhibition restores synaptic transmission and neuronal network activity in a model of chronic stress. Mol Psychiatry 2025; 30:1034-1046. [PMID: 39237722 PMCID: PMC11835755 DOI: 10.1038/s41380-024-02715-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024]
Abstract
Chronic stress exerts profound negative effects on cognitive and emotional behaviours and is a major risk factor for the development of neuropsychiatric disorders. However, the molecular links between chronic stress and its deleterious effects on neuronal and synaptic function remain elusive. Here, using a combination of in vitro and in vivo approaches, we demonstrate that the upregulation of miR-186-5p triggered by chronic stress may be a key mediator of such changes, leading to synaptic dysfunction. Our results show that the expression levels of miR-186-5p are increased both in the prefrontal cortex (PFC) of mice exposed to chronic stress and in cortical neurons chronically exposed to dexamethasone. Additionally, viral overexpression of miR-186-5p in the PFC of naïve mice induces anxiety- and depressive-like behaviours. The upregulation of miR-186-5p through prolonged glucocorticoid receptor activation in vitro, or in a mouse model of chronic stress, differentially affects glutamatergic and GABAergic synaptic transmission, causing an imbalance in excitation/inhibition that leads to altered neuronal network activity. At glutamatergic synapses, we observed both a reduction in synaptic AMPARs and synaptic transmission, whereas GABAergic synaptic transmission was strengthened. These changes could be rescued in vitro by a miR-186-5p inhibitor. Overall, our results establish a novel molecular link between chronic glucocorticoid receptor activation, the upregulation of miR-186-5p and the synaptic changes induced by chronic stress, that may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Beatriz Rodrigues
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- Experimental Biology and Biomedicine Doctoral Programme, Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Ricardo A Leitão
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Mónica Santos
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Alexander Trofimov
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Integrative Brain Function Neurobiology Lab, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, 197022, St. Petersburg, Russia
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, 010000, Astana, Kazakhstan
| | - Mariline Silva
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- Department of Applied Physics and Science for Life Laboratory (SciLifeLab), KTH Royal Institute of Technology, 100 44, Stockholm, Sweden
| | - Ângela S Inácio
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Mónica Abreu
- Multidisciplinary Institute of Aging, MIA Portugal, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Rui J Nobre
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- ViraVector, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Jéssica Costa
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- Experimental Biology and Biomedicine Doctoral Programme, Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Ana Luísa Cardoso
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Ira Milosevic
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Multidisciplinary Institute of Aging, MIA Portugal, University of Coimbra, 3004-504, Coimbra, Portugal
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - João Peça
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456, Coimbra, Portugal
| | - Bárbara Oliveiros
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- iCRB-Coimbra Institute for Clinical and Biomedical Research, University of Coimbra, 3000-548, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- ViraVector, University of Coimbra, 3004-504, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Paulo S Pinheiro
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal.
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456, Coimbra, Portugal.
| | - Ana Luísa Carvalho
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal.
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456, Coimbra, Portugal.
| |
Collapse
|
5
|
Zhang J, Zhong S, Lai S, Zhang Y, Chen G, Huang D, Yan S, Chen P, Lu X, Yin J, Chen C, Wang Y, Jia Y. MIR218 polygenic risk score is associated with cognitive function and neurochemical metabolites among patients with depressed bipolar disorders. J Affect Disord 2025; 371:104-112. [PMID: 39561923 DOI: 10.1016/j.jad.2024.11.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/22/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUNDS Evidence from animal and population studies has consistently revealed that microRNA 218 (MIR218) is involved in susceptibility to depression and cognitive functions. Nevertheless, few studies have evaluated the association between MIR218 and clinical features in patients with depressed bipolar disorder (BD). METHODS A total of 66 patients with depressed BD and 49 healthy controls (HCs) were recruited for this study. MIR218 polygenic risk score (PRS) was used to assess the addictive effects of the MIR218 regulated genes. We compared the MIR218 PRS between patients with depressed BD and HCs to investigate whether it can be used to predict the risk of BD, and further explored the association between MIR218 PRS and cognitive performance as well as neurochemical metabolites among depressed BD. RESULTS We found that there was a significant difference in MIR218 PRS between patients with depressed BD and HCs. The correlation analysis indicated that MIR218 PRS was negative associated with the number of disease onset (r = -0.311, P = 0.033) and choline (Cho)/creatine (Cr) in right thalamus (r = -0.285, P = 0.021). Additionally, as supported by previous findings, patients with lower MIR218 PRS presented more domains of impaired cognitive function than those with higher scores. CONCLUSION These findings suggested MIR218 PRS might be useful in differentiating patients with depressed BD from HCs. Moreover, depressed BD with lower MIR218 PRS showed more pronounced cognitive impairment than those with higher scores, which may be associated with disease recurrence and Cho metabolism in right thalamus.
Collapse
Affiliation(s)
- Jianzhao Zhang
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shuming Zhong
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shunkai Lai
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yiliang Zhang
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Guanmao Chen
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Dong Huang
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shuya Yan
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Pan Chen
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Xiaodan Lu
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jie Yin
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Chao Chen
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410083, China.
| | - Ying Wang
- Medical Imaging Center, First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Yanbin Jia
- Department of Psychiatry, First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| |
Collapse
|
6
|
Cheng P, Ding K, Chen D, Yang C, Wang J, Yang S, Chen M, Zhu G. mPFC DCC coupling with CaMKII + neuronal excitation participates in behavioral despair in male mice. Transl Psychiatry 2025; 15:52. [PMID: 39952936 PMCID: PMC11829057 DOI: 10.1038/s41398-025-03266-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 01/13/2025] [Accepted: 01/30/2025] [Indexed: 02/17/2025] Open
Abstract
A longed lack of control over harmful stimuli can lead to learned helplessness (LH), a significant factor in depression. However, the cellular and molecular mechanisms underlying LH, and eventually behavioral despair, remain largely unknown. The deleted in colorectal cancer (dcc) gene is associated with the risk of depression. However, the therapeutic potential and regulation mechanism of DCC in behavioral despair are still uncertain. In this study, we showed that depressive stimulators, including LH, lipopolysaccharide, and unpredictable chronic mild stress, triggered an elevation in DCC expression in the medial prefrontal cortex (mPFC). Additionally, elevated DCC expression in the mPFC was crucial in inducing behavioral despair, as evidenced by the induction of behavioral despair in normal mice and exacerbation of behavioral despair in LH mice upon DCC overexpression. By contrast, neutralizing DCC activity ameliorated LH-induced behavioral despair. Importantly, we elucidated that pathological DCC expression was attributable to the excessive excitation of CaMKII+ neurons in a manner dependent on the calpain-mediated degradation of SCOP and aberrant phosphorylation of the ERK signaling pathway. In addition, the increase in DCC expression led to a decreased excitability threshold in CaMKII+ neurons in the mPFC, which was supported by the observation that the ligand netrin 1 increased the frequency of action potential firing and of spontaneous excitatory postsynaptic currents in CaMKII+ neurons. In conclusion, our data indicate that LH triggers the excessive excitation of CaMKII+ neurons and activation of calpain-SCOP/ERK signaling to promote DCC expression, and DCC represents a crucial target for the treatment of LH-induced behavioral despair in male mice.
Collapse
Affiliation(s)
- Ping Cheng
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Keke Ding
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Daokang Chen
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Chen Yang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Juan Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Shaojie Yang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Ming Chen
- MOE Frontier Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
| | - Guoqi Zhu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
7
|
Zhong X, Chen Y, Chen W, Liu Y, Gui S, Pu J, Wang D, He Y, Chen X, Chen X, Qiao R, Xie P. Identification of Potential Biomarkers for Major Depressive Disorder: Based on Integrated Bioinformatics and Clinical Validation. Mol Neurobiol 2024; 61:10355-10364. [PMID: 38722514 DOI: 10.1007/s12035-024-04217-1] [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: 03/15/2024] [Accepted: 05/06/2024] [Indexed: 11/24/2024]
Abstract
Major depressive disorder (MDD) is a severe mental illness characterized by a lack of objective biomarkers. Mounting evidence suggests there are extensive transcriptional molecular changes in the prefrontal cortex (PFC) of individuals with MDD. However, it remains unclear whether there are specific genes that are consistently altered and possess diagnostic power. In this study, we conducted a systematic search of PFC datasets of MDD patients from the Gene Expression Omnibus database. We calculated the differential expression of genes (DEGs) and identified robust DEGs using the RRA and MetaDE methods. Furthermore, we validated the consistently altered genes and assessed their diagnostic power through enzyme-linked immunosorbent assay experiments in our clinical blood cohort. Additionally, we evaluated the diagnostic power of hub DEGs in independent public blood datasets. We obtained eight PFC datasets, comprising 158 MDD patients and 263 healthy controls, and identified a total of 1468 unique DEGs. Through integrated analysis, we identified 290 robustly altered DEGs. Among these, seven hub DEGs (SLC1A3, PON2, AQP1, EFEMP1, GJA1, CENPD, HSD11B1) were significantly down-regulated at the protein level in our clinical blood cohort. Moreover, these hub DEGs exhibited a negative correlation with the Hamilton Depression Scale score (P < 0.05). Furthermore, these hub DEGs formed a panel with promising diagnostic power in three independent public blood datasets (average AUCs of 0.85) and our clinical blood cohort (AUC of 0.92). The biomarker panel composed of these genes demonstrated promising diagnostic efficacy for MDD and serves as a useful tool for its diagnosis.
Collapse
Affiliation(s)
- Xiaogang Zhong
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, China
| | - Yue Chen
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, 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
- The Jin Feng Laboratory, Chongqing, 401329, China
- Department of Neurology, 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
- The Jin Feng Laboratory, Chongqing, 401329, China
| | - Siwen Gui
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, 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
- The Jin Feng Laboratory, Chongqing, 401329, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dongfang Wang
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, China
| | - Yong He
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, China
| | - Xiang Chen
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Jin Feng Laboratory, Chongqing, 401329, China
- Department of Neurology, 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
- The Jin Feng Laboratory, Chongqing, 401329, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Renjie Qiao
- 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
| | - Peng Xie
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China.
- NHC Key Laboratory of Diagnosis and Treatment On Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- The Jin Feng Laboratory, Chongqing, 401329, China.
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
8
|
Hu J, Li H, Wang X, Cheng H, Zhu G, Yang S. Novel mechanisms of Anshen Dingzhi prescription against PTSD: Inhibiting DCC to modulate synaptic function and inflammatory responses. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118425. [PMID: 38848974 DOI: 10.1016/j.jep.2024.118425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Anshen Dingzhi prescription (ADP), documented in "Yi Xue Xin Wu", is a famous prescription for treating panic-related mental disorders such as post-traumatic stress disorder (PTSD). However, the underlying mechanism remains unclear. AIM OF THE STUDY This study aimed to investigate the mechanisms by which ADP intervened in PTSD-like behaviors. METHODS A mouse model of single prolonged stress (SPS) was established to evaluate the ameliorative effects and mechanisms of ADP on PTSD. Behavioral tests were used to assess PTSD-like behaviors in mice; transmission electron microscopy was used to observe changes in the ultrastructure of hippocampal synapses, and western blot, immunofluorescence, and ELISA were used to detect the expression of hippocampal deleted in colorectal cancer (DCC) and downstream Ras-related C3 botulinum toxin substrate 1 (Rac1) - P21-activated kinase 1 (PAK1) signal, as well as levels of synaptic proteins and inflammatory factors. Molecular docking technology simulated the binding of potential brain-penetrating components of ADP to DCC. RESULTS SPS induced PTSD-like behaviors in mice and increased expression of hippocampal netrin-1 (NT-1) and DCC on the 14th day post-modeling, with concurrent elevation in serum NT-1 levels. Simultaneously, SPS also decreased p-Rac1 level and increased p-PAK1 level, the down-stream molecules of DCC. Lentiviral overexpression of DCC induced or exacerbated PTSD-like behaviors in control and SPS mice, respectively, whereas neutralization antibody against NT-1 reduced DCC activation and ameliorated PTSD-like behaviors in SPS mice. Interestingly, downstream Rac1-PAK1 signal was altered according to DCC expression. Moreover, DCC overexpression down-regulated N-methyl-d-aspartate (NMDA) receptor 2A (GluN2A) and postsynaptic density 95 (PSD95), up-regulated NMDA receptor 2B (GluN2B) and increased neuroinflammatory responses. Administration of ADP (36.8 mg/kg) improved PTSD-like behaviors in the SPS mice, suppressed hippocampal DCC, and downstream Rac1-PAK1 signal, upregulated GluN2A and PSD95, downregulated GluN2B, and reduced levels of inflammatory factors NOD-like receptor protein 3 (NLRP3), nuclear factor kappa-B (NF-κB) and interleukin-6 (IL-6). Importantly, DCC overexpression could also reduce the ameliorative effect of ADP on PTSD. Additionally, DCC demonstrated a favorable molecular docking pattern with the potential brain-penetrating components of ADP, further suggesting DCC as a potential target of ADP. CONCLUSION Our data indicate that DCC is a key target for the regulation of synaptic function and inflammatory response in the onset of PTSD, and ADP likely reduces DCC to prevent PTSD via modulating downstream Rac1-PAK1 pathway. This study provides a novel mechanism for the onset of PTSD and warrants the clinical application of ADP.
Collapse
Affiliation(s)
- Jiamin Hu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China.
| | - Haipeng Li
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China.
| | - Xuncui Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China.
| | - Hongliang Cheng
- The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, 230061, China.
| | - Guoqi Zhu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China.
| | - Shaojie Yang
- The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, 230061, China.
| |
Collapse
|
9
|
Dong T, Yu C, Mao Q, Han F, Yang Z, Yang Z, Pires N, Wei X, Jing W, Lin Q, Hu F, Hu X, Zhao L, Jiang Z. Advances in biosensors for major depressive disorder diagnostic biomarkers. Biosens Bioelectron 2024; 258:116291. [PMID: 38735080 DOI: 10.1016/j.bios.2024.116291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/25/2024] [Accepted: 04/09/2024] [Indexed: 05/14/2024]
Abstract
Depression is one of the most common mental disorders and is mainly characterized by low mood or lack of interest and pleasure. It can be accompanied by varying degrees of cognitive and behavioral changes and may lead to suicide risk in severe cases. Due to the subjectivity of diagnostic methods and the complexity of patients' conditions, the diagnosis of major depressive disorder (MDD) has always been a difficult problem in psychiatry. With the discovery of more diagnostic biomarkers associated with MDD in recent years, especially emerging non-coding RNAs (ncRNAs), it is possible to quantify the condition of patients with mental illness based on biomarker levels. Point-of-care biosensors have emerged due to their advantages of convenient sampling, rapid detection, miniaturization, and portability. After summarizing the pathogenesis of MDD, representative biomarkers, including proteins, hormones, and RNAs, are discussed. Furthermore, we analyzed recent advances in biosensors for detecting various types of biomarkers of MDD, highlighting representative electrochemical sensors. Future trends in terms of new biomarkers, new sample processing methods, and new detection modalities are expected to provide a complete reference for psychiatrists and biomedical engineers.
Collapse
Affiliation(s)
- Tao Dong
- X Multidisciplinary Research Institute, School of Instrument Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China; Chongqing Key Laboratory of Micro-Nano Transduction and Intelligent Systems, Collaborative Innovation Center on Micro-Nano Transduction and Intelligent Eco-Internet of Things, Chongqing Key Laboratory of Colleges and Universities on Micro-Nano Systems Technology and Smart Transducing, National Research Base of Intelligent Manufacturing Service, Chongqing Technology and Business University, Nan'an District, Chongqing, 400067, China.
| | - Chenghui Yu
- Chongqing Key Laboratory of Micro-Nano Transduction and Intelligent Systems, Collaborative Innovation Center on Micro-Nano Transduction and Intelligent Eco-Internet of Things, Chongqing Key Laboratory of Colleges and Universities on Micro-Nano Systems Technology and Smart Transducing, National Research Base of Intelligent Manufacturing Service, Chongqing Technology and Business University, Nan'an District, Chongqing, 400067, China.
| | - Qi Mao
- X Multidisciplinary Research Institute, School of Instrument Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Feng Han
- X Multidisciplinary Research Institute, School of Instrument Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhenwei Yang
- X Multidisciplinary Research Institute, School of Instrument Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhaochu Yang
- Chongqing Key Laboratory of Micro-Nano Transduction and Intelligent Systems, Collaborative Innovation Center on Micro-Nano Transduction and Intelligent Eco-Internet of Things, Chongqing Key Laboratory of Colleges and Universities on Micro-Nano Systems Technology and Smart Transducing, National Research Base of Intelligent Manufacturing Service, Chongqing Technology and Business University, Nan'an District, Chongqing, 400067, China
| | - Nuno Pires
- Chongqing Key Laboratory of Micro-Nano Transduction and Intelligent Systems, Collaborative Innovation Center on Micro-Nano Transduction and Intelligent Eco-Internet of Things, Chongqing Key Laboratory of Colleges and Universities on Micro-Nano Systems Technology and Smart Transducing, National Research Base of Intelligent Manufacturing Service, Chongqing Technology and Business University, Nan'an District, Chongqing, 400067, China
| | - Xueyong Wei
- X Multidisciplinary Research Institute, School of Instrument Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Weixuan Jing
- X Multidisciplinary Research Institute, School of Instrument Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Qijing Lin
- X Multidisciplinary Research Institute, School of Instrument Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Fei Hu
- X Multidisciplinary Research Institute, School of Instrument Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xiao Hu
- Engineering Research Center of Ministry of Education for Smart Justice, School of Criminal Investigation, Southwest University of Political Science and Law, Chongqing, 401120, China.
| | - Libo Zhao
- X Multidisciplinary Research Institute, School of Instrument Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhuangde Jiang
- X Multidisciplinary Research Institute, School of Instrument Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
10
|
Morgunova A, Teixeira M, Flores C. Perspective on adolescent psychiatric illness and emerging role of microRNAs as biomarkers of risk. J Psychiatry Neurosci 2024; 49:E282-E288. [PMID: 39209460 PMCID: PMC11374446 DOI: 10.1503/jpn.240072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Affiliation(s)
- Alice Morgunova
- From the Douglas Mental Health University Institute, Montreal, Que. (Morgunova, Flores); the Department of Psychiatry, McGill University, Montreal, Que. (Morgunova, Flores); the Integrated Program in Neuroscience, McGill University, Montreal, Que. (Teixeira); the Department of Neurology and Neurosurgery, McGill University, Montreal, Que. (Flores); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montreal, Que. (Flores)
| | - Maxime Teixeira
- From the Douglas Mental Health University Institute, Montreal, Que. (Morgunova, Flores); the Department of Psychiatry, McGill University, Montreal, Que. (Morgunova, Flores); the Integrated Program in Neuroscience, McGill University, Montreal, Que. (Teixeira); the Department of Neurology and Neurosurgery, McGill University, Montreal, Que. (Flores); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montreal, Que. (Flores)
| | - Cecilia Flores
- From the Douglas Mental Health University Institute, Montreal, Que. (Morgunova, Flores); the Department of Psychiatry, McGill University, Montreal, Que. (Morgunova, Flores); the Integrated Program in Neuroscience, McGill University, Montreal, Que. (Teixeira); the Department of Neurology and Neurosurgery, McGill University, Montreal, Que. (Flores); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montreal, Que. (Flores)
| |
Collapse
|
11
|
Chen HS, Wang F, Chen JG. Epigenetic mechanisms in depression: Implications for pathogenesis and treatment. Curr Opin Neurobiol 2024; 85:102854. [PMID: 38401316 DOI: 10.1016/j.conb.2024.102854] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/03/2024] [Accepted: 02/05/2024] [Indexed: 02/26/2024]
Abstract
The risk of depression is influenced by both genetic and environmental factors. It has been suggested that epigenetic mechanisms may mediate the risk of depression following exposure to adverse life events. Epigenetics encompasses stable alterations in gene expression that are controlled through transcriptional, post-transcriptional, translational, or post-translational processes, including DNA modifications, chromatin remodeling, histone modifications, RNA modifications, and non-coding RNA (ncRNA) regulation, without any changes in the DNA sequence. In this review, we explore recent research advancements in the realm of epigenetics concerning depression. Furthermore, we evaluate the potential of epigenetic changes as diagnostic and therapeutic biomarkers for depression.
Collapse
Affiliation(s)
- Hong-Sheng Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China; The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan 430030, China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China; The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan 430030, China; The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan 430030, China
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China; The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan 430030, China; The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan 430030, China.
| |
Collapse
|
12
|
Musazzi L, Mingardi J, Ieraci A, Barbon A, Popoli M. Stress, microRNAs, and stress-related psychiatric disorders: an overview. Mol Psychiatry 2023; 28:4977-4994. [PMID: 37391530 DOI: 10.1038/s41380-023-02139-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/23/2023] [Accepted: 06/16/2023] [Indexed: 07/02/2023]
Abstract
Stress is a major risk factor for psychiatric disorders. During and after exposure to stressors, the stress response may have pro- or maladaptive consequences, depending on several factors related to the individual response and nature of the stressor. However, the mechanisms mediating the long-term effects of exposure to stress, which may ultimately lead to the development of stress-related disorders, are still largely unknown. Epigenetic mechanisms have been shown to mediate the effects of the environment on brain gene expression and behavior. MicroRNAs, small non-coding RNAs estimated to control the expression of about 60% of all genes by post-transcriptional regulation, are a fundamental epigenetic mechanism. Many microRNAs are expressed in the brain, where they work as fine-tuners of gene expression, with a key role in the regulation of homeostatic balance, and a likely influence on pro- or maladaptive brain changes. Here we have selected a number of microRNAs, which have been strongly implicated as mediators of the effects of stress in the brain and in the development of stress-related psychiatric disorders. For all of them recent evidence is reported, obtained from rodent stress models, manipulation of microRNAs levels with related behavioral changes, and clinical studies of stress-related psychiatric disorders. Moreover, we have performed a bioinformatic analysis of the predicted brain-expressed target genes of the microRNAs discussed, and found a central role for mechanisms involved in the regulation of synaptic function. The complex regulatory role of microRNAs has suggested their use as biomarkers for diagnosis and treatment response, as well as possible therapeutic drugs. While, microRNA-based diagnostics have registered advancements, particularly in oncology and other fields, and many biotech companies have launched miRNA therapeutics in their development pipeline, the development of microRNA-based tests and drugs for brain disorders is comparatively slower.
Collapse
Affiliation(s)
- Laura Musazzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Jessica Mingardi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Alessandro Ieraci
- Department of Theoretical and Applied Sciences, eCampus University, Novedrate, Italy
- Molecular Pharmacology, Cellular and Behavioral Physiology; Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy
| | - Alessandro Barbon
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy.
| |
Collapse
|
13
|
Taylor SR, Kobayashi M, Vilella A, Tiwari D, Zolboot N, Du JX, Spencer KR, Hartzell A, Girgiss C, Abaci YT, Shao Y, De Sanctis C, Bellenchi GC, Darnell RB, Gross C, Zoli M, Berg DK, Lippi G. MicroRNA-218 instructs proper assembly of hippocampal networks. eLife 2023; 12:e82729. [PMID: 37862092 PMCID: PMC10637775 DOI: 10.7554/elife.82729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/10/2023] [Indexed: 10/21/2023] Open
Abstract
The assembly of the mammalian brain is orchestrated by temporally coordinated waves of gene expression. Post-transcriptional regulation by microRNAs (miRNAs) is a key aspect of this program. Indeed, deletion of neuron-enriched miRNAs induces strong developmental phenotypes, and miRNA levels are altered in patients with neurodevelopmental disorders. However, the mechanisms used by miRNAs to instruct brain development remain largely unexplored. Here, we identified miR-218 as a critical regulator of hippocampal assembly. MiR-218 is highly expressed in the hippocampus and enriched in both excitatory principal neurons (PNs) and GABAergic inhibitory interneurons (INs). Early life inhibition of miR-218 results in an adult brain with a predisposition to seizures. Changes in gene expression in the absence of miR-218 suggest that network assembly is impaired. Indeed, we find that miR-218 inhibition results in the disruption of early depolarizing GABAergic signaling, structural defects in dendritic spines, and altered intrinsic membrane excitability. Conditional knockout of Mir218-2 in INs, but not PNs, is sufficient to recapitulate long-term instability. Finally, de-repressing Kif21b and Syt13, two miR-218 targets, phenocopies the effects on early synchronous network activity induced by miR-218 inhibition. Taken together, the data suggest that miR-218 orchestrates formative events in PNs and INs to produce stable networks.
Collapse
Affiliation(s)
- Seth R Taylor
- Division of Biological Sciences, University of California, San DiegoLa JollaUnited States
| | - Mariko Kobayashi
- Laboratory of Molecular Neuro-oncology, Howard Hughes Medical Institute, Rockefeller UniversityNew YorkUnited States
| | - Antonietta Vilella
- Department of Biomedical, Metabolic and Neural Sciences; Center for Neuroscience and Neurotechnology (CfNN), University of Modena and Reggio EmiliaModenaItaly
| | - Durgesh Tiwari
- Division of Neurology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of MedicineCincinnatiUnited States
- Department of Pediatrics, University of Cincinnati College of MedicineCincinnatiUnited States
| | - Norjin Zolboot
- Department of Neuroscience, Scripps Research InstituteLa JollaUnited States
| | - Jessica X Du
- Department of Neuroscience, Scripps Research InstituteLa JollaUnited States
| | - Kathryn R Spencer
- Department of Neuroscience, Scripps Research InstituteLa JollaUnited States
| | - Andrea Hartzell
- Department of Neuroscience, Scripps Research InstituteLa JollaUnited States
| | - Carol Girgiss
- Division of Biological Sciences, University of California, San DiegoLa JollaUnited States
| | - Yusuf T Abaci
- Division of Biological Sciences, University of California, San DiegoLa JollaUnited States
| | - Yufeng Shao
- Department of Neuroscience, Scripps Research InstituteLa JollaUnited States
| | | | - Gian Carlo Bellenchi
- Institute of Genetics and Biophysics A Buzzati-TraversoNaplesItaly
- IRCCS Fondazione Santa LuciaRomeItaly
| | - Robert B Darnell
- Laboratory of Molecular Neuro-oncology, Howard Hughes Medical Institute, Rockefeller UniversityNew YorkUnited States
| | - Christina Gross
- Division of Neurology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of MedicineCincinnatiUnited States
- Department of Pediatrics, University of Cincinnati College of MedicineCincinnatiUnited States
| | - Michele Zoli
- Department of Biomedical, Metabolic and Neural Sciences; Center for Neuroscience and Neurotechnology (CfNN), University of Modena and Reggio EmiliaModenaItaly
| | - Darwin K Berg
- Division of Biological Sciences, University of California, San DiegoLa JollaUnited States
| | - Giordano Lippi
- Department of Neuroscience, Scripps Research InstituteLa JollaUnited States
| |
Collapse
|
14
|
Batra A, Cuesta S, Alves MB, Restrepo JM, Giroux M, Laureano DP, Mucellini Lovato AB, Miguel PM, Machado TD, Molle RD, Flores C, Silveira PP. Relationship between insulin and Netrin-1/DCC guidance cue pathway regulation in the prefrontal cortex of rodents exposed to prenatal dietary restriction. J Dev Orig Health Dis 2023; 14:501-507. [PMID: 37431265 PMCID: PMC10988268 DOI: 10.1017/s204017442300017x] [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] [Indexed: 07/12/2023]
Abstract
Fetal restriction (FR) alters insulin sensitivity, but it is unknown how the metabolic profile associated with restriction affects development of the dopamine (DA) system and DA-related behaviors. The Netrin-1/DCC guidance cue system participates in maturation of the mesocorticolimbic DA circuitry. Therefore, our objective was to identify if FR modifies Netrin-1/DCC receptor protein expression in the prefrontal cortex (PFC) at birth and mRNA in adulthood in rodent males. We used cultured HEK293 cells to assess if levels of miR-218, microRNA regulator of DCC, are sensitive to insulin. To assess this, pregnant dams were subjected to a 50% FR diet from gestational day 10 until birth. Medial PFC (mPFC) DCC/Netrin-1 protein expression was measured at P0 at baseline and Dcc/Netrin-1 mRNA levels were quantified in adults 15 min after a saline/insulin injection. miR-218 levels in HEK-293 cells were measured in response to insulin exposure. At P0, Netrin-1 levels are downregulated in FR animals in comparison to controls. In adult rodents, insulin administration results in an increase in Dcc mRNA levels in control but not FR rats. In HEK293 cells, there is a positive correlation between insulin concentration and miR-218 levels. Since miR-218 is a Dcc gene expression regulator and our in vitro results show that insulin regulates miR-218 levels, we suggest that FR-induced changes in insulin sensitivity could be affecting Dcc expression via miR-218, impacting DA system maturation and organization. As fetal adversity is linked to nonadaptive behaviors later in life, this may contribute to early identification of vulnerability to chronic diseases associated with fetal adversity.
Collapse
Affiliation(s)
- Aashita Batra
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Santiago Cuesta
- Department of Cell Biology and Neuroscience, Rutgers University, New Brunswick, NJ, USA
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Marcio Bonesso Alves
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Jose Maria Restrepo
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Michel Giroux
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Daniela Pereira Laureano
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Amanda Brondani Mucellini Lovato
- Programa de Pós-Graduação em Psiquiatria e Ciências do Comportamento, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Patrícia Maidana Miguel
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Tania Diniz Machado
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Roberta Dalle Molle
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Cecilia Flores
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Patricia Pelufo Silveira
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
15
|
Jiang T, Yang Y, Wu C, Qu C, Chen JG, Cao H. MicroRNA-218 regulates neuronal radial migration and morphogenesis by targeting Satb2 in developing neocortex. Biochem Biophys Res Commun 2023; 647:9-15. [PMID: 36708662 DOI: 10.1016/j.bbrc.2023.01.053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023]
Abstract
Neuronal migration and morphogenesis are fundamental processes for cortical development. Their defects may cause abnormities in neural circuit formation and even neuropsychiatric disorders. Many proteins, especially layer-specific transcription factors and adhesion molecules, have been reported to regulate the processes. However, the involvement of non-coding RNAs in cortical development has not been extensively studied. Here, we identified microRNA-218 (miR-218) as a layer V-specific microRNA in mouse brains. Expression of miR-218 was elevated in patients with autism spectrum disorder (ASD) and schizophrenia. We found in this study that miR-218 overexpression in developing mouse cortex led to severe defects in radial migration, morphogenesis, and spatial distribution of the cortical neurons. Moreover, we identified Satb2, an upper-layer marker, as a molecular target repressed by miR-218. These results suggest an underlying mechanism of miR-218 involvement in neuropsychiatric disorders, and the interactions of layer-specific non-coding RNAs and proteins in regulating cortical development.
Collapse
Affiliation(s)
- Tian Jiang
- Department of Clinical Laboratory, The Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, 317500, PR China; School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, State Key Laboratory of Optometry, Ophthalmology and Vision Science, and Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, PR China
| | - Yaojuan Yang
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, State Key Laboratory of Optometry, Ophthalmology and Vision Science, and Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, PR China
| | - Chunping Wu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, State Key Laboratory of Optometry, Ophthalmology and Vision Science, and Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, PR China
| | - Chunsheng Qu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, State Key Laboratory of Optometry, Ophthalmology and Vision Science, and Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, PR China
| | - Jie-Guang Chen
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, State Key Laboratory of Optometry, Ophthalmology and Vision Science, and Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, PR China.
| | - Huateng Cao
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, State Key Laboratory of Optometry, Ophthalmology and Vision Science, and Zhejiang Provincial Key Laboratory of Optometry and Ophthalmology, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, PR China.
| |
Collapse
|
16
|
Roy B, Dwivedi Y. An insight into the sprawling microverse of microRNAs in depression pathophysiology and treatment response. Neurosci Biobehav Rev 2023; 146:105040. [PMID: 36639069 PMCID: PMC9974865 DOI: 10.1016/j.neubiorev.2023.105040] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/12/2023]
Abstract
Stress-related neuropathologies are pivotal in developing major depressive disorder (MDD) and are often governed by gene-regulatory changes. Being a stress-responsive gene-regulatory factor, microRNAs (miRNAs) have tremendous biomolecular potential to define an altered gene-regulatory landscape in the MDD brain. MiRNAs' regulatory roles in the MDD brain are closely aligned with changes in plasticity, neurogenesis, and stress-axis functions. MiRNAs act at the epigenetic interface between stress-induced environmental stimuli and cellular pathologies by triggering large-scale gene expression changes in a highly coordinated fashion. The parallel changes in peripheral circulation may provide an excellent opportunity for miRNA to devise more effective treatment strategies and help explore their potential as biomarkers in treatment response. This review discusses the role of miRNAs as epigenetic modifiers in the etiopathogenesis of MDD. Concurrently, key research is highlighted to show the progress in using miRNAs as predictive biomarkers for treatment response.
Collapse
Affiliation(s)
- Bhaskar Roy
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
17
|
Potential of Circulating miRNAs as Molecular Markers in Mood Disorders and Associated Suicidal Behavior. Int J Mol Sci 2023; 24:ijms24054664. [PMID: 36902096 PMCID: PMC10003208 DOI: 10.3390/ijms24054664] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/23/2023] [Accepted: 02/26/2023] [Indexed: 03/04/2023] Open
Abstract
Mood disorders are the most prevalent psychiatric disorders associated with significant disability, morbidity, and mortality. The risk of suicide is associated with severe or mixed depressive episodes in patients with mood disorders. However, the risk of suicide increases with the severity of depressive episodes and is often presented with higher incidences in bipolar disorder (BD) patients than in patients with major depression (MDD). Biomarker study in neuropsychiatric disorders is critical for developing better treatment plans by facilitating more accurate diagnosis. At the same time, biomarker discovery also provides more objectivity to develop state-of-the-art personalized medicine with increased accuracy through clinical interventions. Recently, colinear changes in miRNA expression between brain and systemic circulation have added great interest in examining their potential as molecular markers in mental disorders, including MDD, BD, and suicidality. A present understanding of circulating miRNAs in body fluids implicates their role in managing neuropsychiatric conditions. Most notably, their use as prognostic and diagnostic markers and their potential role in treatment response have significantly advanced our knowledge base. The present review discusses circulatory miRNAs and their underlying possibilities to be used as a screening tool for assessing major psychiatric conditions, including MDD, BD, and suicidal behavior.
Collapse
|
18
|
Popa N, Bachar D, Roberts AC, Santangelo AM, Gascon E. Region-specific microRNA alterations in marmosets carrying SLC6A4 polymorphisms are associated with anxiety-like behavior. EBioMedicine 2022; 82:104159. [PMID: 35905539 PMCID: PMC9334339 DOI: 10.1016/j.ebiom.2022.104159] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 06/21/2022] [Accepted: 06/28/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Psychiatric diseases such as depression and anxiety are multifactorial conditions, highly prevalent in western societies. Human studies have identified a number of high-risk genetic variants for these diseases. Among them, polymorphisms in the promoter region of the serotonin transporter gene (SLC6A4) have attracted much attention. However, due to the paucity of experimental models, molecular alterations induced by these genetic variants and how they correlate to behavioral deficits have not been examined. In this regard, marmosets have emerged as a powerful model in translational neuroscience to investigate molecular underpinnings of complex behaviors. METHODS Here, we took advantage of naturally occurring genetic polymorphisms in marmoset SLC6A4 gene that have been linked to anxiety-like behaviors. Using FACS-sorting, we profiled microRNA contents in different brain regions of genotyped and behaviorally-phenotyped marmosets. FINDINGS We revealed that marmosets bearing different SLC6A4 variants exhibit distinct microRNAs signatures in a region of the prefrontal cortex whose activity has been consistently altered in patients with depression/anxiety. We also identified Deleted in Colorectal Cancer (DCC), a gene previously linked to these diseases, as a downstream target of the differently expressed microRNAs. Significantly, we showed that levels of both microRNAs and DCC in this region were highly correlated to anxiety-like behaviors. INTERPRETATION Our findings establish links between genetic variants, molecular modifications in specific cortical regions and complex behavioral responses, providing new insights into gene-behavior relationships underlying human psychopathology. FUNDING This work was supported by France National Agency, NRJ Foundation, Celphedia and Fondation de France as well as the Wellcome Trust.
Collapse
Affiliation(s)
- Natalia Popa
- Aix Marseille Univ, CNRS, INT, Inst Neurosci Timone, Marseille, France
| | - Dipankar Bachar
- Aix Marseille Univ, CNRS, INT, Inst Neurosci Timone, Marseille, France
| | - Angela C Roberts
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Andrea M Santangelo
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Eduardo Gascon
- Aix Marseille Univ, CNRS, INT, Inst Neurosci Timone, Marseille, France.
| |
Collapse
|
19
|
Vázquez-Ágredos A, Gámiz F, Gallo M. MicroRNA Regulation of the Environmental Impact on Adolescent Neurobehavioral Development: A Systematic Review. Front Cell Neurosci 2022; 16:956609. [PMID: 35936504 PMCID: PMC9352948 DOI: 10.3389/fncel.2022.956609] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Adolescence is a late developmental period marked by pronounced reorganization of brain networks in which epigenetic mechanisms play a fundamental role. This brain remodeling is associated with a peculiar behavior characterized by novelty seeking and risky activities such as alcohol and drug abuse, which is associated with increased susceptibility to stress. Hence, adolescence is a vulnerable postnatal period since short- and long-term deleterious effects of alcohol drinking and drug abuse are a serious worldwide public health concern. Among several other consequences, it has been proposed that exposure to stress, alcohol, or other drugs disrupts epigenetic mechanisms mediated by small non-coding microRNAs (miRNAs). During adolescence, this modifies the expression of a variety of genes involved in neurodevelopmental processes such as proliferation, differentiation, synaptogenesis, neural plasticity, and apoptosis. Hence, the effect of miRNAs dysregulation during adolescence might contribute to a long-term impact on brain function. This systematic review focuses on the miRNA expression patterns in the adolescent rodent brain with special interest in the impact of stress and drugs such as amphetamine, cocaine, nicotine, cannabis, and ketamine. The results point to a relevant and complex role of miRNAs in the regulation of the molecular processes involved in adolescent brain development as part of a dynamic epigenetic network sensitive to environmental events with distinctive changes across adolescence. Several miRNAs have been assessed evidencing changing expression profiles during the adolescent transition which are altered by exposure to stress and drug abuse. Since this is an emerging rapidly growing field, updating the present knowledge will contribute to improving our understanding of the epigenetic regulation mechanisms involved in the neurodevelopmental changes responsible for adolescent behavior. It can be expected that increased knowledge of the molecular mechanisms mediating the effect of environmental threats during the adolescent critical developmental period will improve understanding of psychiatric and addictive disorders emerging at this stage.
Collapse
Affiliation(s)
- Ana Vázquez-Ágredos
- Department of Psychobiology, Institute of Neurosciences (CIBM), University of Granada, Granada, Spain
| | - Fernando Gámiz
- Department of Psychobiology, Institute of Neurosciences (CIBM), University of Granada, Granada, Spain
| | - Milagros Gallo
- Department of Psychobiology, Institute of Neurosciences (CIBM), University of Granada, Granada, Spain
| |
Collapse
|
20
|
Schell G, Roy B, Prall K, Dwivedi Y. miR-218: A Stress-Responsive Epigenetic Modifier. Noncoding RNA 2022; 8:ncrna8040055. [PMID: 35893238 PMCID: PMC9326663 DOI: 10.3390/ncrna8040055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022] Open
Abstract
Understanding the epigenetic role of microRNAs (miRNAs) has been a critical development in the field of neuropsychiatry and in understanding their underlying pathophysiology. Abnormalities in miRNA expression are often seen as key to the pathogenesis of many stress-associated mental disorders, including major depressive disorder (MDD). Recent advances in omics biology have further contributed to this understanding and expanded the role of miRNAs in networking a diverse array of molecular pathways, which are essentially related to the stress adaptivity of a healthy brain. Studies have highlighted the role of many such miRNAs in causing maladaptive changes in the brain's stress axis. One such miRNA is miR-218, which is debated as a critical candidate for increased stress susceptibility. miR-218 is expressed throughout the brain, notably in the hippocampus and prefrontal cortex (PFC). It is expressed at various levels through life stages, as seen by adolescent and adult animal models. Until now, a minimal number of studies have been conducted on human subjects to understand its role in stress-related abnormalities in brain circuits. However, several studies, including animal and cell-culture models, have been used to understand the impact of miR-218 on stress response and hypothalamic-pituitary-adrenal (HPA) axis function. So far, expression changes in this miRNA have been found to regulate signaling pathways such as glucocorticoid signaling, serotonergic signaling, and glutamatergic signaling. Recently, the developmental role of miR-218 has generated interest, given its increasing expression from adolescence to adulthood and targeting the Netrin-1/DCC signaling pathway. Since miR-218 expression affects neuronal development and plasticity, it is expected that a change in miR-218 expression levels over the course of development may negatively impact the process and make individuals stress-susceptible in adulthood. In this review, we describe the role of miR-218 in stress-induced neuropsychiatric conditions with an emphasis on stress-related disorders.
Collapse
|
21
|
Next RNA Therapeutics: The Mine of Non-Coding. Int J Mol Sci 2022; 23:ijms23137471. [PMID: 35806476 PMCID: PMC9267739 DOI: 10.3390/ijms23137471] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/02/2022] [Accepted: 07/04/2022] [Indexed: 12/26/2022] Open
Abstract
The growing knowledge on several classes of non-coding RNAs (ncRNAs) and their different functional roles has aroused great interest in the scientific community. Beyond the Central Dogma of Biology, it is clearly known that not all RNAs code for protein products, and they exert a broader repertoire of biological functions. As described in this review, ncRNAs participate in gene expression regulation both at transcriptional and post-transcriptional levels and represent critical elements driving and controlling pathophysiological processes in multicellular organisms. For this reason, in recent years, a great boost was given to ncRNA-based strategies with potential therapeutic abilities, and nowadays, the use of RNA molecules is experimentally validated and actually exploited in clinics to counteract several diseases. In this review, we summarize the principal classes of therapeutic ncRNA molecules that are potentially implied in disease onset and progression, which are already used in clinics or under clinical trials, highlighting the advantages and the need for a targeted therapeutic strategy design. Furthermore, we discuss the benefits and the limits of RNA therapeutics and the ongoing development of delivery strategies to limit the off-target effects and to increase the translational application.
Collapse
|
22
|
Zhou S, Chen R, She Y, Liu X, Zhao H, Li C, Jia Y. A new perspective on depression and neuroinflammation: Non-coding RNA. J Psychiatr Res 2022; 148:293-306. [PMID: 35193033 DOI: 10.1016/j.jpsychires.2022.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 01/30/2022] [Accepted: 02/14/2022] [Indexed: 12/19/2022]
Abstract
The high incidence and relapse rate of depression, as well comorbidity with other diseases, has made depression one of the primary causes of years of life lived with disability. Moreover, the unknown biological mechanism of depression has made treatment difficult. Neuroinflammation is important in the pathogenesis of depression. Neuroinflammation may affect depression by regulating the production of immune factors, immune cell activation, neuron generation, synaptic plasticity, and neurotransmission. Non-coding RNAs (ncRNAs) may be a breakthrough link between depression and neuroinflammation, as ncRNAs participate in these biological changes. We summarize the functions and mechanisms of ncRNAs in neuroinflammation and depression, and predict ncRNAs that may regulate the occurrence and progression of depression through neuritis. These findings not only broaden our understanding of the genetic regulation of depression and neuroinflammation but also provide a new perspective of the underlying mechanism and aid in the design of novel prevention, diagnosis, and treatment strategies.
Collapse
Affiliation(s)
- Shanyao Zhou
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, 466 Xin Gang Zhong Road, Guangzhou, 510317, China
| | - Rui Chen
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, 466 Xin Gang Zhong Road, Guangzhou, 510317, China.
| | - Yanling She
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, 466 Xin Gang Zhong Road, Guangzhou, 510317, China
| | - Xuanjun Liu
- Department of Psychiatry, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Guangzhou, 510630, China
| | - Hui Zhao
- Department of Psychiatry, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Guangzhou, 510630, China
| | - Cheng Li
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, 466 Xin Gang Zhong Road, Guangzhou, 510317, China.
| | - Yanbin Jia
- Department of Psychiatry, First Affiliated Hospital of Jinan University, 613 W. Huangpu Avenue, Guangzhou, 510630, China.
| |
Collapse
|
23
|
Elias E, Zhang AY, Manners MT. Novel Pharmacological Approaches to the Treatment of Depression. Life (Basel) 2022; 12:196. [PMID: 35207483 PMCID: PMC8879976 DOI: 10.3390/life12020196] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/19/2022] [Accepted: 01/23/2022] [Indexed: 12/18/2022] Open
Abstract
Major depressive disorder is one of the most prevalent mental health disorders. Monoamine-based antidepressants were the first drugs developed to treat major depressive disorder. More recently, ketamine and other analogues were introduced as fast-acting antidepressants. Unfortunately, currently available therapeutics are inadequate; lack of efficacy, adverse effects, and risks leave patients with limited treatment options. Efforts are now focused on understanding the etiology of depression and identifying novel targets for pharmacological treatment. In this review, we discuss promising novel pharmacological targets for the treatment of major depressive disorder. Targeting receptors including N-methyl-D-aspartate receptors, peroxisome proliferator-activated receptors, G-protein-coupled receptor 39, metabotropic glutamate receptors, galanin and opioid receptors has potential antidepressant effects. Compounds targeting biological processes: inflammation, the hypothalamic-pituitary-adrenal axis, the cholesterol biosynthesis pathway, and gut microbiota have also shown therapeutic potential. Additionally, natural products including plants, herbs, and fatty acids improved depressive symptoms and behaviors. In this review, a brief history of clinically available antidepressants will be provided, with a primary focus on novel pharmaceutical approaches with promising antidepressant effects in preclinical and clinical studies.
Collapse
Affiliation(s)
| | | | - Melissa T. Manners
- Department of Biological Sciences, University of the Sciences, 600 South 43rd Street, Philadelphia, PA 19104, USA; (E.E.); (A.Y.Z.)
| |
Collapse
|
24
|
Morgunova A, Flores C. MicroRNAs as promising peripheral sensors of prefrontal cortex developmental trajectory and psychiatric risk. Neuropsychopharmacology 2022; 47:387-388. [PMID: 34312494 PMCID: PMC8617183 DOI: 10.1038/s41386-021-01113-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Alice Morgunova
- Integrated Program in Neuroscience (IPN), McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Cecilia Flores
- Douglas Mental Health University Institute, Montreal, QC, Canada.
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC, Canada.
- Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
| |
Collapse
|
25
|
Zhou H, Xiang W, Huang M. Inactivation of Zona Incerta Blocks Social Conditioned Place Aversion and Modulates Post-traumatic Stress Disorder-Like Behaviors in Mice. Front Behav Neurosci 2021; 15:743484. [PMID: 34744654 PMCID: PMC8568071 DOI: 10.3389/fnbeh.2021.743484] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/29/2021] [Indexed: 11/19/2022] Open
Abstract
Zona incerta (ZI), a largely inhibitory subthalamic region connected with many brain areas, has been suggested to serve as an integrative node for modulation of behaviors and physiological states, such as fear memory conditioning and aversion responses. It is, however, unclear whether ZI regulated the repeated social defeat stress (RSDS)-induced social conditioned place aversion (CPA) and post-traumatic stress disorder (PTSD)-like behaviors. In this study, the function of ZI was silenced via bilateral injection of tetanus toxin light chain (Tet-tox), a neurotoxin that completely blocks the evoked synaptic transmissions, expressing adeno-associated viruses (AAVs). We found ZI silencing: (1) significantly blocked the expression of RSDS-induced social CPA with no effect on the innate preference; (2) significantly enhanced the anxiety level in mice experienced RSDS with no effect on the locomotion activity; (3) altered the PTSD-associated behaviors, including the promotion of spatial cognitive impairment and the preventions of PPI deficit and social avoidance behavior. These effects were not observed on non-stressed mice. In summary, our results suggest the important role of ZI in modulating RSDS-induced social CPA and PTSD-like behaviors.
Collapse
Affiliation(s)
- Hong Zhou
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Xiang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengbing Huang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Reynolds LM, Flores C. Mesocorticolimbic Dopamine Pathways Across Adolescence: Diversity in Development. Front Neural Circuits 2021; 15:735625. [PMID: 34566584 PMCID: PMC8456011 DOI: 10.3389/fncir.2021.735625] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/17/2021] [Indexed: 12/26/2022] Open
Abstract
Mesocorticolimbic dopamine circuity undergoes a protracted maturation during adolescent life. Stable adult levels of behavioral functioning in reward, motivational, and cognitive domains are established as these pathways are refined, however, their extended developmental window also leaves them vulnerable to perturbation by environmental factors. In this review, we highlight recent advances in understanding the mechanisms underlying dopamine pathway development in the adolescent brain, and how the environment influences these processes to establish or disrupt neurocircuit diversity. We further integrate these recent studies into the larger historical framework of anatomical and neurochemical changes occurring during adolescence in the mesocorticolimbic dopamine system. While dopamine neuron heterogeneity is increasingly appreciated at molecular, physiological, and anatomical levels, we suggest that a developmental facet may play a key role in establishing vulnerability or resilience to environmental stimuli and experience in distinct dopamine circuits, shifting the balance between healthy brain development and susceptibility to psychiatric disease.
Collapse
Affiliation(s)
- Lauren M Reynolds
- Plasticité du Cerveau CNRS UMR8249, École supérieure de physique et de chimie industrielles de la Ville de Paris (ESPCI Paris), Paris, France.,Neuroscience Paris Seine CNRS UMR 8246 INSERM U1130, Institut de Biologie Paris Seine, Sorbonne Université, Paris, France
| | - Cecilia Flores
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| |
Collapse
|