1
|
Lakhawat SS, Mech P, Kumar A, Malik N, Kumar V, Sharma V, Bhatti JS, Jaswal S, Kumar S, Sharma PK. Intricate mechanism of anxiety disorder, recognizing the potential role of gut microbiota and therapeutic interventions. Metab Brain Dis 2024; 40:64. [PMID: 39671133 DOI: 10.1007/s11011-024-01453-1] [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: 02/21/2024] [Accepted: 09/29/2024] [Indexed: 12/14/2024]
Abstract
Anxiety is a widespread psychological disorder affecting both humans and animals. It is a typical stress reaction; however, its longer persistence can cause severe health disorders affecting the day-to-day life activities of individuals. An intriguing facet of the anxiety-related disorder can be addressed better by investigating the role of neurotransmitters in regulating emotions, provoking anxiety, analyzing the cross-talks between neurotransmitters, and, most importantly, identifying the biomarkers of the anxiety. Recent years have witnessed the potential role of the gut microbiota in human health and disorders, including anxiety. Animal models are commonly used to study anxiety disorder as they offer a simpler and more controlled environment than humans. Ultimately, developing new strategies for diagnosing and treating anxiety is of paramount interest to medical scientists. Altogether, this review article shall highlight the intricate mechanisms of anxiety while emphasizing the emerging role of gut microbiota in regulating metabolic pathways through various interaction networks in the host. In addition, the review will foster information about the therapeutic interventions of the anxiety and related disorder.
Collapse
Affiliation(s)
- Sudarshan Singh Lakhawat
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India
| | - Priyanka Mech
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India
| | - Akhilesh Kumar
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India
| | - Naveen Malik
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India
| | - Vikram Kumar
- Amity Institute of Pharmacy, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, India
| | - Vinay Sharma
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India
| | - Jasvinder Singh Bhatti
- Department of Environmental Sciences, Himachal Pradesh University, Summer Hill, Shimla, 171005, India
| | - Sunil Jaswal
- Department of Human Genetics and Molecular Medicine Central University Punjab, Bathinda, 151401, India
| | - Sunil Kumar
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India
| | - Pushpender Kumar Sharma
- Amity Institute of Biotechnology, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India.
- Amity Centre for Nanobiotechnology and Nanomedicine, Amity University Rajasthan, SP-1, Kant Kalwar, RIICO Industrial Area, NH-11C, Jaipur, Rajasthan, 303002, India.
| |
Collapse
|
2
|
Cai Y, Zhao R, Huang Y, Yang H, Liu Y, Yang R, Zhang X, Liu Y, Yan S, Liu X, Liu X, Yin X, Yu Y, Gao S, Li Y, Zhao Y, Shi H. Environmental enrichment attenuates maternal separation-induced excessive hoarding behavior in adult female mice. Pharmacol Biochem Behav 2024; 245:173913. [PMID: 39581387 DOI: 10.1016/j.pbb.2024.173913] [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: 08/20/2024] [Revised: 11/17/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Previous studies have demonstrated that early life stress (ELS) impacts hoarding behavior in adult humans. This study aimed to assess the potential mitigation by environmental enrichment on hoarding behavior in rodents caused by maternal separation, thereby providing insights into therapeutic strategies for hoarding disorder. METHODS Newborn mice were randomly divided into four groups. The control group was allowed to grow naturally. The maternal separation group underwent two weeks of maternal separation. The short-term environmental enrichment group received two weeks of environmental enrichment intervention after the two weeks of maternal separation. The long-term environmental enrichment group received five weeks of environmental enrichment intervention after the two weeks of maternal separation. Hoarding behavior was assessed during adolescence and adulthood. Hippocampal tissue from adult female mice was analyzed using LC-MS/MS-based metabolomics. Spearman correlation analysis was then performed to assess the relationship between differentially expressed metabolites and hoarding behavior. RESULTS Environmental enrichment attenuates maternal separation-induced excessive hoarding behavior in adult female mice. The untargeted metabolomics of the hippocampal region in female mice showed that long-term environmental enrichment reversed multiple differential metabolites, including Substance P, which were mainly concentrated in metabolic pathways such as cancer choline metabolism, glycolipid metabolism, and linoleic acid metabolism. CONCLUSIONS Our findings indicate that ELS and long-term environmental enrichment have sex-dependent effects on adult hoarding behavior, potentially related to altered hippocampal metabolism. This study highlights the importance of environmental enrichment in mitigating the long-term effects of early maternal separation on hoarding behavior.
Collapse
Affiliation(s)
- Yiming Cai
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Ruofan Zhao
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Yuxuan Huang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Huiping Yang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Ye Liu
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Rui Yang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiangyu Zhang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Yiran Liu
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Shu Yan
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiaoyu Liu
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiao Liu
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Xueyong Yin
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Yang Yu
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Shuai Gao
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Yating Li
- Nursing School, Hebei Medical University, Shijiazhuang 050031, China
| | - Ye Zhao
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang 050017, China.
| | - Haishui Shi
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Nursing School, Hebei Medical University, Shijiazhuang 050031, China; Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang 050017, China; Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
3
|
Yang D, Bai R, Li C, Sun Y, Jing H, Wang Z, Chen Y, Dong Y. Early-Life Stress Induced by Neonatal Maternal Separation Leads to Intestinal 5-HT Accumulation and Causes Intestinal Dysfunction. J Inflamm Res 2024; 17:8945-8964. [PMID: 39588137 PMCID: PMC11586501 DOI: 10.2147/jir.s488290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024] Open
Abstract
Background The early childhood period is a critical development stage, and experiencing stress during this time may increase the risk of gastrointestinal disorders, including irritable bowel syndrome (IBS). Neonatal maternal separation (NMS) in rodent models has been shown to cause bowel dysfunctions similar to IBS, and 5-HT is considered to be a key regulator regulating intestinal function, but the precise underlying mechanisms remain unclear. Results We established a maternal separation stress mouse model to simulate early-life stress, exploring the expression patterns of 5-HT under chronic stress and its mechanisms affecting gut function. We observed a significant increase in 5-HT expression due to NMS, leading to disruptions in intestinal structure and function. However, inhibiting 5-HT reversed these effects, suggesting its potential as a therapeutic target. Furthermore, our research revealed that excess 5-HT in mice with early life stress increased intestinal neural network density and promoted excitatory motor neuron expression. Mechanistically, 5-HT activated the Wnt signaling pathway through the 5-HT4 receptor, promoting neurogenesis within the intestinal nervous system. Conclusion These findings shed light on the intricate changes induced by early life stress in the intestines, confirming the regulatory role of 5-HT in the enteric nervous system and providing potential insights for the development of novel therapies for gastrointestinal disorders.
Collapse
Affiliation(s)
- Ding Yang
- College of Veterinary Medicine, China Agricultural University, Beijing, People’s Republic of China
| | - Rulan Bai
- College of Veterinary Medicine, China Agricultural University, Beijing, People’s Republic of China
| | - Chengzhong Li
- Department of Horticulture and Landscape Architecture, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, People’s Republic of China
| | - Yan Sun
- Department of Horticulture and Landscape Architecture, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, People’s Republic of China
| | - Hongyu Jing
- College of Veterinary Medicine, China Agricultural University, Beijing, People’s Republic of China
| | - Zixu Wang
- College of Veterinary Medicine, China Agricultural University, Beijing, People’s Republic of China
| | - Yaoxing Chen
- College of Veterinary Medicine, China Agricultural University, Beijing, People’s Republic of China
| | - Yulan Dong
- College of Veterinary Medicine, China Agricultural University, Beijing, People’s Republic of China
| |
Collapse
|
4
|
Chen CY, Wang YF, Lei L, Zhang Y. MicroRNA-specific targets for neuronal plasticity, neurotransmitters, neurotrophic factors, and gut microbes in the pathogenesis and therapeutics of depression. Prog Neuropsychopharmacol Biol Psychiatry 2024; 136:111186. [PMID: 39521033 DOI: 10.1016/j.pnpbp.2024.111186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/11/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Depression is of great concern because of the huge burden, and it is impacted by various epigenetic modifications, e.g., histone modification, covalent modifications in DNA, and silencing mechanisms of non-coding protein genes, e.g., microRNAs (miRNAs). MiRNAs are a class of endogenous non-coding RNAs. Alternations in specific miRNAs have been observed both in depressive patients and experimental animals. Also, miRNAs are highly expressed in the central nervous system and can be delivered to different tissues via tissue-specific exosomes. However, the mechanism of miRNAs' involvement in the pathological process of depression is not well understood. Therefore, we summarized and discussed the role of miRNAs in depression. Conclusively, miRNAs are involved in the pathology of depression by causing structural and functional changes in synapses, mediating neuronal regeneration, differentiation, and apoptosis, regulating the gut microbes and the expression of various neurotransmitters and BDNF, and mediating inflammatory and immune responses. Moreover, miRNAs can predict the efficacy of antidepressant medications and explain the mechanism of action of antidepressant drugs and aerobic exercise to prevent and assist in treating depression.
Collapse
Affiliation(s)
- Cong-Ya Chen
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yu-Fei Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Lan Lei
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
5
|
Schmidt AT, Hicks SD, Bergquist BK, Maloney KA, Dennis VE, Bammel AC. Preliminary Evidence for Neuronal Dysfunction Following Adverse Childhood Experiences: An Investigation of Salivary MicroRNA Within a High-Risk Youth Sample. Genes (Basel) 2024; 15:1433. [PMID: 39596633 PMCID: PMC11593590 DOI: 10.3390/genes15111433] [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: 09/26/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Adverse childhood experiences (ACEs) are potent drivers of psychopathology and neurological disorders, especially within minoritized populations. Nonetheless, we lack a coherent understanding of the neuronal mechanisms through which ACEs impact gene expression and, thereby, the development of psychopathology. Methods: This observational pilot study used a novel marker of neuronal functioning (brain-derived micro ribonucleic acids, or miRNAs) collected via saliva to explore the connection between ACEs and neuronal gene expression in 45 adolescents with a collectively high ACE exposure (26 males and 19 females of diverse races/ethnicities, with six cumulative ACEs on average). We aimed to determine the feasibility of using salivary microRNA for probing neuronal gene expression with the goal of identifying cellular processes and genetic pathways perturbed by childhood adversity. Results: A total of 274 miRNAs exhibited reliable salivary expression (raw counts > 10 in > 10% of samples). Fourteen (5.1%) were associated with cumulative ACE exposure (p < 0.05; r's ≥ 0.31). ACE exposure correlated negatively with miR-92b-3p, 145a-5p, 31-5p, and 3065-5p, and positively with miR-15b-5p, 30b-5p, 30c-5p, 30e-3p, 199a-3p, 223-3p, 338-3p, 338-5p, 542-3p, and 582-5p. Most relations remained significant after controlling for multiple comparisons and potential retrospective bias in ACE reporting for miRNAs with particularly strong relations (p < 0.03). We examined KEGG pathways targeted by miRNAs associated with total ACE scores. Results indicated putative miRNA targets over-represented 47 KEGG pathways (adjusted p < 0.05) involved in neuronal signaling, brain development, and neuroinflammation. Conclusions: Although preliminary and with a small sample, the findings represent a novel contribution to the understanding of how childhood adversity impacts neuronal gene expression via miRNA signaling.
Collapse
Affiliation(s)
- Adam T. Schmidt
- Department of Psychological Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Center for Translational Neuroscience and Therapeutics, TTUHSC, Lubbock, TX 79409, USA
| | - Steven D. Hicks
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA 17033, USA;
| | - Becca K. Bergquist
- Department of Psychological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Kelsey A. Maloney
- Department of Pediatrics, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Victoria E. Dennis
- Department of Psychological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Alexandra C. Bammel
- Department of Psychological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
6
|
Qiu X, Danesh Yazdi M, Wang C, Kosheleva A, Wu H, Vokonas PS, Spiro A, Laurent LC, DeHoff P, Kubzansky LD, Weisskopf MG, Baccarelli AA, Schwartz JD. Extracellular microRNAs associated with psychiatric symptoms in the Normative Aging Study. J Psychiatr Res 2024; 178:270-277. [PMID: 39173451 DOI: 10.1016/j.jpsychires.2024.08.017] [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: 03/07/2024] [Revised: 06/15/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024]
Abstract
Earlier studies have revealed microRNAs (miRNAs) as potential biomarkers for neurological conditions, however, such evidence on psychiatric outcomes is limited. We utilized the Normative Aging Study (NAS) cohort to investigate the associations between extracellular miRNAs (ex-miRNA) and psychiatric symptoms among a group of older male adults, along with the targeted genes and biological pathways. We studied 569 participants with miRNA profile primarily measured in extracellular vesicles isolated from plasma, and psychiatric symptoms reported over 1996-2014 with repeated measures. Global and dimension scales of psychiatric symptoms were measured via the administration of Brief Symptom Inventory (BSI) per visit covering nine aspects of psychiatric health, such as anxiety, depression, hostility, psychoticism, etc. Ex-miRNAs were profiled using small RNA sequencing. Associations of expression of 395 ex-miRNAs (present in >70% samples) with current mental status were assessed using single-miRNA as well as Least Absolute Shrinkage and Selection Operator (LASSO)-based multi-miRNAs linear mixed effects models adjusting for key demographic and behavioral factors. Biological functions were explored using pathway analyses. We identified ex-miRNAs associated with each BSI scale. In particular, hsa-miR-320d was consistently identified for two global scales. Similar overlapping miRNAs across global and dimension scores included hsa-miR-379-3p, hsa-miR-1976, hsa-miR-151a-5p, hsa-miR-151b, hsa-miR-144-3p, etc. Top KEGG pathways for identified miRNAs included p53 signaling, Hippo signaling, FoxO signaling, protein processing in endoplasmic reticulum and several pathways related with cancer and neurological diseases. This study provided early evidence supporting the associations between extracellular miRNAs and psychiatric conditions. MiRNAs may serve as biomarkers of subclinical psychiatric illness in older adults.
Collapse
Affiliation(s)
- Xinye Qiu
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Mahdieh Danesh Yazdi
- Program in Public Health, Department of Family, Population, and Preventive Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Cuicui Wang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Anna Kosheleva
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Haotian Wu
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Pantel S Vokonas
- Veterans Affairs (VA) Normative Aging Study, VA Boston Healthcare System, Boston, MA, USA; Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Avron Spiro
- Veterans Affairs (VA) Normative Aging Study, VA Boston Healthcare System, Boston, MA, USA; Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA; Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Louise C Laurent
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Peter DeHoff
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Laura D Kubzansky
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Marc G Weisskopf
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andrea A Baccarelli
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Joel D Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
7
|
Benatti BM, Adiletta A, Sgadò P, Malgaroli A, Ferro M, Lamanna J. Epigenetic Modifications and Neuroplasticity in the Pathogenesis of Depression: A Focus on Early Life Stress. Behav Sci (Basel) 2024; 14:882. [PMID: 39457754 PMCID: PMC11504006 DOI: 10.3390/bs14100882] [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: 07/18/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Major depressive disorder (MDD) is a debilitating mental illness, and it is considered to be one of the leading causes of disability globally. The etiology of MDD is multifactorial, involving an interplay between biological, psychological, and social factors. Early life represents a critical period for development. Exposure to adverse childhood experiences is a major contributor to the global burden of disease and disability, doubling the risk of developing MDD later in life. Evidence suggests that stressful events experienced during that timeframe play a major role in the emergence of MDD, leading to epigenetic modifications, which might, in turn, influence brain structure, function, and behavior. Neuroplasticity seems to be a primary pathogenetic mechanism of MDD, and, similarly to epigenetic mechanisms, it is particularly sensitive to stress in the early postnatal period. In this review, we will collect and discuss recent studies supporting the role of epigenetics and neuroplasticity in the pathogenesis of MDD, with a focus on early life stress (ELS). We believe that understanding the epigenetic mechanisms by which ELS affects neuroplasticity offers potential pathways for identifying novel therapeutic targets for MDD, ultimately aiming to improve treatment outcomes for this debilitating disorder.
Collapse
Affiliation(s)
- Bianca Maria Benatti
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, 20132 Milan, Italy; (B.M.B.); (M.F.)
| | - Alice Adiletta
- Center for Mind/Brain Sciences, University of Trento, 38068 Rovereto, Italy; (A.A.); (P.S.)
| | - Paola Sgadò
- Center for Mind/Brain Sciences, University of Trento, 38068 Rovereto, Italy; (A.A.); (P.S.)
| | - Antonio Malgaroli
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, 20132 Milan, Italy; (B.M.B.); (M.F.)
- Faculty of Psychology, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Clinical Center Tourette Syndrome, IRCCS Ospedale San Raffaele, 20127 Milan, Italy
| | - Mattia Ferro
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, 20132 Milan, Italy; (B.M.B.); (M.F.)
- Department of Psychology, Sigmund Freud Private University, 20143 Milan, Italy
| | - Jacopo Lamanna
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, 20132 Milan, Italy; (B.M.B.); (M.F.)
- Clinical Center Tourette Syndrome, IRCCS Ospedale San Raffaele, 20127 Milan, Italy
| |
Collapse
|
8
|
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 2024:10.1038/s41380-024-02715-1. [PMID: 39237722 DOI: 10.1038/s41380-024-02715-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
9
|
Liu X, He C, Bai Y, Fan D, Zang F, Zhang H, Zhang H, Yao H, Zhang Z, Xie C. White matter microstructure mediates the pairwise relationship between childhood maltreatment, microRNA-9, and the severity of major depressive disorder. Chin Med J (Engl) 2024; 137:2140-2142. [PMID: 39113182 PMCID: PMC11374261 DOI: 10.1097/cm9.0000000000003188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Indexed: 09/06/2024] Open
Affiliation(s)
- Xinyi Liu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Cancan He
- Neuropsychiatric Research Institute, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Ying Bai
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Dandan Fan
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
- Neuropsychiatric Research Institute, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Feifei Zang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
- Neuropsychiatric Research Institute, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Hongxing Zhang
- Henan Key Laboratory of Psychology and Behavior, Xinxiang Medical University, Xinxiang, Henan 453000, China
- Department of Radiology, Henan Provincial Mental Hospital, Xinxiang Medical University, Xinxiang, Henan 453000, China
| | - Haisan Zhang
- Psychology School of Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Honghong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
- Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210009, China
| | - Zhijun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
- Neuropsychiatric Research Institute, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
- Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210009, China
| | - Chunming Xie
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
- Neuropsychiatric Research Institute, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
- Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
10
|
Pereira SC, Coeli-Lacchini FB, Pereira DA, Ferezin LP, Menezes IC, Baes CVW, Luizon MR, Juruena MF, Cleare AJ, Young AH, Lacchini R. Early life stress unravels epistatic genetic associations of cortisol pathway genes with depression. J Psychiatr Res 2024; 175:323-332. [PMID: 38759498 DOI: 10.1016/j.jpsychires.2024.05.032] [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: 01/26/2024] [Revised: 03/26/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024]
Abstract
Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis represents one of the most consistent pathophysiological findings in depressive disorders. Cortisol signaling is affected by proteins that mediate its cellular responses or alters its availability to mineralocorticoid and glucocorticoid receptors. In our study, we evaluated candidate genes that may influence the risk for depression and suicide due to its involvement in cortisol signaling. The aim of the study was to assess whether the genotypes of these genes are associated with the risk for depression, severity of depressive symptoms, suicidal ideation, and suicide attempts. And whether there is interaction between genes and early-life stress. In this study, 100 healthy controls and 140 individuals with depression were included. The subjects were clinically assessed using the 21-item GRID-Hamilton questionnaires (GRID-HAMD-21), Beck Scale for Suicidal Ideation (BSI), and the Childhood Trauma Questionnaire (CTQ). A robust multifactorial dimensionality reduction analysis was used to characterize the interactions between the genes HSD11B1, NR3C1, NR3C2, and MDR1 and early-life stress. It was found a significant association of the heterozygous genotype of the MDR1 gene rs1128503 polymorphism with reduced risk of at least one suicide attempt (OR: 0.08, p = 0.003*) and a reduction in the number of suicide attempts (β = -0.79, p = 0.006*). Furthermore, it was found that the MDR1 rs1228503 and NR3C2 rs2070951 genes interact with early-life stress resulting in a strong association with depression (p = 0.001). Our findings suggest that polymorphisms in the MDR1 and NR3C2 genes and their interaction with childhood trauma may be important biomarkers for depression and suicidal behaviors.
Collapse
Affiliation(s)
- Sherliane Carla Pereira
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | - Daniela Alves Pereira
- Department of Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Letícia Perticarrara Ferezin
- Department of Public Health Nursing, Ribeirão Preto Nursing School, University of São Paulo, Ribeirão Preto, Brazil
| | - Itiana Castro Menezes
- Department of Neuroscience and Behavior, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Cristiane von Werne Baes
- Department of Neuroscience and Behavior, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Marcelo Rizzatti Luizon
- Department of Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mario F Juruena
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London & South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road, Beckenham, Kent, BR3 3BX, United Kingdom
| | - Anthony J Cleare
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London & South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road, Beckenham, Kent, BR3 3BX, United Kingdom
| | - Allan H Young
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London & South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road, Beckenham, Kent, BR3 3BX, United Kingdom
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, Brazil.
| |
Collapse
|
11
|
Zhang Y, Gong L, Feng Q, Hu K, Liu C, Jiang T, Zhang Q. Association between negative life events through mental health and non-suicidal self-injury with young adults: evidence for sex moderate correlation. BMC Psychiatry 2024; 24:466. [PMID: 38914977 PMCID: PMC11197180 DOI: 10.1186/s12888-024-05880-3] [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: 09/22/2023] [Accepted: 05/31/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Non-suicidal self-injury (NSSI) has exhibited an increasing trend in recent years and is now globally recognized as a major public health problem among adolescents and young adults. Negative life events (NLEs) are positively associated with NSSI. We sought to explore (1) whether sex plays a role in the risk of NLEs leading to NSSI and (2) the role played by mental health (MH). METHODS We adopted a multi-stage cluster sampling method to select college students across four grades from May to June 2022. Generalized linear models were used to evaluate the relationships between NLEs, sex, MH and NSSI, presented as incidence-rate ratios (RRs) with 95% confidence intervals (CIs). We examined the complex relationship between these variables using the PROCESS method for moderation analysis. RESULTS Following the exclusion of data that did not meet the study requirements, data from 3,578 students (mean age: 20.53 [± 1.65] years) were included. Poisson regression results indicate that high-level NLEs (RR = 0.110, 95%CI: 0.047-0.173) are associated with increased NSSI. Furthermore, interaction effects were observed among sex, NLEs and NSSI. MH and sex moderated the relationship between NLEs and NSSI. CONCLUSION Identifying risk factors for NSSI is also important when exploring the interaction between NLEs and MH given the potential for NSSI to significantly increase the risk of later psychopathological symptoms and substance abuse problems. In addition, the significance of sex differences in risk factors for NSSI should be determined. This study evaluated how the impact of NLEs on NSSI can be reduced among adolescents from multiple perspectives.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Li Gong
- Wuxi Huishan District People's Hospital, Wuxi, Jiangsu, 214187, China
| | - Qing Feng
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Keyan Hu
- The First Affiliated Hospital, College of Clinical Medicine of Henan, University of Science and Technology, Luoyang, Henan, 471003, China
| | - Chao Liu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Tian Jiang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China.
| | - Qiu Zhang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China.
| |
Collapse
|
12
|
Yin X, Zhao Y, Wang S, Feng H, He X, Li X, Liu X, Lu H, Wen D, Shi Y, Shi H. Postweaning stress affects behavior, brain and gut microbiota of adolescent mice in a sex-dependent manner. Neuropharmacology 2024; 248:109869. [PMID: 38354850 DOI: 10.1016/j.neuropharm.2024.109869] [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: 10/17/2023] [Revised: 01/29/2024] [Accepted: 02/10/2024] [Indexed: 02/16/2024]
Abstract
Aggression is an instinctive behavior that has been reported to be influenced by early-life stress. However, the potential effects of acute stress during the postweaning period, a key stage for brain development, on defensive aggression and the associated mechanism remain poorly understood. In the present study, aggressive behaviors were evaluated in adolescent mice exposed to postweaning stress. Serum corticosterone and testosterone levels, neural dendritic spine density, and gut microbiota composition were determined to identify the underlying mechanism. Behavioral analysis showed that postweaning stress reduced locomotor activity in mice and decreased defensive aggression in male mice. ELISA results showed that postweaning stress reduced serum testosterone levels in female mice. Golgi staining analysis demonstrated that postweaning stress decreased neural dendritic spine density in the medial prefrontal cortex of male mice. 16S rRNA sequencing results indicated that postweaning stress altered the composition of the gut microbiota in male mice. Combined, these results suggested that postweaning stress alters defensive aggression in male mice, which may be due to changes in neuronal structure as well as gut microbiota composition. Our findings highlight the long-lasting and sex-dependent effects of early-life experience on behaviors.
Collapse
Affiliation(s)
- Xueyong Yin
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Ye Zhao
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shuang Wang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Hao Feng
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xinyue He
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xincheng Li
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xiaoyu Liu
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Hengtai Lu
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Di Wen
- Hebei Key Laboratory of Forensic Medicine, Hebei Province, Shijiazhuang, 050017, China
| | - Yun Shi
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Haishui Shi
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Forensic Medicine, Hebei Province, Shijiazhuang, 050017, China; Nursing School, Hebei Medical University, Shijiazhuang, 050031, China.
| |
Collapse
|
13
|
Bai Y, Shu C, Hou Y, Wang GH. Adverse childhood experience and depression: the role of gut microbiota. Front Psychiatry 2024; 15:1309022. [PMID: 38628262 PMCID: PMC11019508 DOI: 10.3389/fpsyt.2024.1309022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Depression is the most common psychiatric disorder that burdens modern society heavily. Numerous studies have shown that adverse childhood experiences can increase susceptibility to depression, and depression with adverse childhood experiences has specific clinical-biological features. However, the specific neurobiological mechanisms are not yet precise. Recent studies suggest that the gut microbiota can influence brain function and behavior associated with depression through the "microbe-gut-brain axis" and that the composition and function of the gut microbiota are influenced by early stress. These studies offer a possibility that gut microbiota mediates the relationship between adverse childhood experiences and depression. However, few studies directly link adverse childhood experiences, gut microbiota, and depression. This article reviews recent studies on the relationship among adverse childhood experiences, gut microbiota, and depression, intending to provide insights for new research.
Collapse
Affiliation(s)
- Yu Bai
- Department of Psychiatry, Renmin Hospital of Wuhan University, Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chang Shu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ying Hou
- Peking University China-Japan Friendship School of Clinical Medicine, Department of Neurology, Beijing, China
| | - Gao-Hua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
14
|
Zhang L, Xie Q, Chang S, Ai Y, Dong K, Zhang H. Epigenetic Factor MicroRNAs Likely Mediate Vaccine Protection Efficacy against Lymphomas in Response to Tumor Virus Infection in Chickens through Target Gene Involved Signaling Pathways. Vet Sci 2024; 11:139. [PMID: 38668407 PMCID: PMC11053969 DOI: 10.3390/vetsci11040139] [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: 02/21/2024] [Revised: 03/16/2024] [Accepted: 03/20/2024] [Indexed: 04/29/2024] Open
Abstract
Epigenetic factors, including microRNAs (miRNAs), play an important role in affecting gene expression and, therefore, are involved in various biological processes including immunity protection against tumors. Marek's disease (MD) is a highly contagious disease of chickens caused by the MD virus (MDV). MD has been primarily controlled by vaccinations. MD vaccine efficacy might, in part, be dependent on modulations of a complex set of factors including host epigenetic factors. This study was designed to identify differentially expressed miRNAs in the primary lymphoid organ, bursae of Fabricius, in response to MD vaccination followed by MDV challenge in two genetically divergent inbred lines of White Leghorns. Small RNA sequencing and bioinformatic analyses of the small RNA sequence reads identified hundreds of miRNAs among all the treatment groups. A small portion of the identified miRNAs was differentially expressed within each of the four treatment groups, which were HVT or CVI988/Rispens vaccinated line 63-resistant birds and line 72-susceptible birds. A direct comparison between the resistant line 63 and susceptible line 72 groups vaccinated with HVT followed by MDV challenge identified five differentially expressed miRNAs. Gene Ontology analysis of the target genes of those five miRNAs revealed that those target genes, in addition to various GO terms, are involved in multiple signaling pathways including MAPK, TGF-β, ErbB, and EGFR1 signaling pathways. The general functions of those pathways reportedly play important roles in oncogenesis, anti-cancer immunity, cancer cell migration, and metastatic progression. Therefore, it is highly likely that those miRNAs may, in part, influence vaccine protection through the pathways.
Collapse
Affiliation(s)
- Lei Zhang
- U.S. Department of Agriculture, Agricultural Research Service, U.S. National Poultry Research Center, Athens, GA 30605, USA;
- Institute of Special Wild Economic Animal and Plant Science, Chinese Academy of Agricultural Sciences, Changchun 130112, China
| | - Qingmei Xie
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China;
| | - Shuang Chang
- College of Veterinary Medicine, Shandong Agricultural University, Tai’an 271018, China;
| | - Yongxing Ai
- College of Animal Science, Jilin University, Changchun 130062, China;
| | - Kunzhe Dong
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA 30912, USA;
| | - Huanmin Zhang
- U.S. Department of Agriculture, Agricultural Research Service, U.S. National Poultry Research Center, Athens, GA 30605, USA;
| |
Collapse
|
15
|
Reemst K, Lopizzo N, Abbink MR, Engelenburg HJ, Cattaneo A, Korosi A. Molecular underpinnings of programming by early-life stress and the protective effects of early dietary ω6/ω3 ratio, basally and in response to LPS: Integrated mRNA-miRNAs approach. Brain Behav Immun 2024; 117:283-297. [PMID: 38242369 DOI: 10.1016/j.bbi.2024.01.011] [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: 06/02/2023] [Revised: 12/22/2023] [Accepted: 01/14/2024] [Indexed: 01/21/2024] Open
Abstract
Early-life stress (ELS) exposure increases the risk for mental disorders, including cognitive impairments later in life. We have previously demonstrated that an early diet with low ω6/ω3 polyunsaturated fatty acid (PUFA) ratio protects against ELS-induced cognitive impairments. Several studies have implicated the neuroimmune system in the ELS and diet mediated effects, but currently the molecular pathways via which ELS and early diet exert their long-term impact are not yet fully understood. Here we study the effects of ELS and dietary PUFA ratio on hippocampal mRNA and miRNA expression in adulthood, both under basal as well as inflammatory conditions. Male mice were exposed to chronic ELS by the limiting bedding and nesting material paradigm from postnatal day(P)2 to P9, and provided with a diet containing a standard (high (15:1.1)) or protective (low (1.1:1)) ω6 linoleic acid to ω3 alpha-linolenic acid ratio from P2 to P42. At P120, memory was assessed using the object location task. Subsequently, a single lipopolysaccharide (LPS) injection was given and 24 h later hippocampal genome-wide mRNA and microRNA (miRNA) expression was measured using microarray. Spatial learning deficits induced by ELS in mice fed the standard (high ω6/ω3) diet were reversed by the early-life protective (low ω6/ω3) diet. An integrated miRNA - mRNA analysis revealed that ELS and early diet induced miRNA driven mRNA expression changes into adulthood. Under basal conditions both ELS and the diet affected molecular pathways related to hippocampal plasticity, with the protective (low ω6/ω3 ratio) diet leading to activation of molecular pathways associated with improved hippocampal plasticity and learning and memory in mice previously exposed to ELS (e.g., CREB signaling and endocannabinoid neuronal synapse pathway). LPS induced miRNA and mRNA expression was strongly dependent on both ELS and early diet. In mice fed the standard (high ω6/ω3) diet, LPS increased miRNA expression leading to activation of inflammatory pathways. In contrast, in mice fed the protective diet, LPS reduced miRNA expression and altered target mRNA expression inhibiting inflammatory signaling pathways and pathways associated with hippocampal plasticity, which was especially apparent in mice previously exposed to ELS. This data provides molecular insights into how the protective (low ω6/ω3) diet during development could exert its long-lasting beneficial effects on hippocampal plasticity and learning and memory especially in a vulnerable population exposed to stress early in life, providing the basis for the development of intervention strategies.
Collapse
Affiliation(s)
- Kitty Reemst
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science park 904, Amsterdam, 1098 XH, the Netherlands
| | - Nicola Lopizzo
- Biological Psychiatry Unit, Istituto di Recupero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Maralinde R Abbink
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science park 904, Amsterdam, 1098 XH, the Netherlands
| | - Hendrik J Engelenburg
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science park 904, Amsterdam, 1098 XH, the Netherlands
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, Istituto di Recupero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Aniko Korosi
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science park 904, Amsterdam, 1098 XH, the Netherlands.
| |
Collapse
|
16
|
Yu S, Zhao Y, Luo Q, Gu B, Wang X, Cheng J, Wang Z, Liu D, Ho RCM, Ho CSH. Early life stress enhances the susceptibility to depression and interferes with neuroplasticity in the hippocampus of adolescent mice via regulating miR-34c-5p/SYT1 axis. J Psychiatr Res 2024; 170:262-276. [PMID: 38181539 DOI: 10.1016/j.jpsychires.2023.12.030] [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: 09/02/2023] [Revised: 11/14/2023] [Accepted: 12/18/2023] [Indexed: 01/07/2024]
Abstract
Early life events are major risk factors for the onset of depression and have long-term effects on the neurobiological changes and behavioral development of rodents. However, little is known about the specific mechanisms of early life adversity in the susceptibility to subsequent stress exposure in adolescence. This study characterized the effect of maternal separation (MS), an animal model of early life adversity, on the behavioral responses to restraint stress in mice during adolescence and investigated the molecular mechanism underlying behavioral vulnerability to chronic stress induced by MS. Our results showed that MS exposure could further reinforce the depressive vulnerability to restraint stress in adolescent mice. In addition, miR-34c-5p expression was obviously up-regulated in the hippocampi of MS mice at postnatal day (P) 14 and P42. Further, synaptotagmin-1 (SYT1) was deemed as a target gene candidate of miR-34c-5p on the basis of dual luciferase assay. It was found that the downregulation of miR-34c-5p expression in the hippocampi of MS mice could ameliorate dysfunction of synaptic plasticity by targeting molecule SYT1, effects which were accompanied by alleviation of depressive and anxious behaviors in these mice. The results demonstrated that the miR-34c-5p/SYT1 pathway was involved in the susceptibility to depression induced by MS via regulating neuroplasticity in the hippocampi of mice.
Collapse
Affiliation(s)
- Shuwen Yu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yijing Zhao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Qian Luo
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Bing Gu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xixi Wang
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Jiao Cheng
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China.
| | - Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China.
| | - Roger C M Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Institute of Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore
| | - Cyrus S H Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
17
|
Gao Y, Nie K, Wang H, Dong H, Tang Y. Research progress on antidepressant effects and mechanisms of berberine. Front Pharmacol 2024; 15:1331440. [PMID: 38318145 PMCID: PMC10839030 DOI: 10.3389/fphar.2024.1331440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/11/2024] [Indexed: 02/07/2024] Open
Abstract
Depression, a global health problem with growing prevalence, brings serious impacts on the daily life of patients. However, the antidepressants currently used in clinical are not perfectly effective, which greatly reduces the compliance of patients. Berberine is a natural quaternary alkaloid which has been shown to have a variety of pharmacological effects, such as hypoglycemic, lipid-regulation, anti-cancer, antibacterial, anti-oxidation, anti-inflammatory, and antidepressant. This review summarizes the evidence of pharmacological applications of berberine in treating depression and elucidates the mechanisms of berberine regulating neurotransmitter levels, promoting the regeneration of hippocampal neurons, improving hypothalamic-pituitary-adrenal axis dysfunction, anti-oxidative stress, and suppressing inflammatory status in order to provide a reference for further research and clinical application of berberine.
Collapse
Affiliation(s)
- Yang Gao
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kexin Nie
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongzhan Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Dong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yueheng Tang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
18
|
Su H, Zhu L, Su L, Li M, Wang R, Zhu J, Chen Y, Chen T. Impact of miR-29c-3p in the Nucleus Accumbens on Methamphetamine-Induced Behavioral Sensitization and Neuroplasticity-Related Proteins. Int J Mol Sci 2024; 25:942. [PMID: 38256016 PMCID: PMC10815255 DOI: 10.3390/ijms25020942] [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: 11/17/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Methamphetamine (METH) abuse inflicts both physical and psychological harm. While our previous research has established the regulatory role of miR-29c-3p in behavior sensitization, the underlying mechanisms and target genes remain incompletely understood. In this study, we employed the isobaric tags for relative and absolute quantitation (iTRAQ) technique in conjunction with Ingenuity pathway analysis (IPA) to probe the putative molecular mechanisms of METH sensitization through miR-29c-3p inhibition. Through a microinjection of AAV-anti-miR-29c-3p into the nucleus accumbens (NAc) of mice, we observed the attenuation of METH-induced locomotor effects. Subsequent iTRAQ analysis identified 70 differentially expressed proteins (DEPs), with 22 up-regulated potential target proteins identified through miR-29c-3p target gene prediction and IPA analysis. Our focus extended to the number of neuronal branches, the excitatory synapse count, and locomotion-related pathways. Notably, GPR37, NPC1, and IREB2 emerged as potential target molecules for miR-29c-3p regulation, suggesting their involvement in the modulation of METH sensitization. Quantitative PCR confirmed the METH-induced aberrant expression of Gpr37, Npc1, and Ireb2 in the NAc of mice. Specifically, the over-expression of miR-29c-3p led to a significant reduction in the mRNA level of Gpr37, while the inhibition of miR-29c-3p resulted in a significant increase in the mRNA level of Gpr37, consistent with the regulatory principle of miRNAs modulating target gene expression. This suggests that miR-29c-3p potentially influences METH sensitization through its regulation of neuroplasticity. Our research indicates that miR-29c-3p plays a crucial role in regulating METH-induced sensitization, and it identified the potential molecular of miR-29c-3p in regulating METH-induced sensitization.
Collapse
Affiliation(s)
- Hang Su
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (H.S.); (L.Z.); (L.S.); (M.L.); (R.W.); (J.Z.)
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Xi’an 710061, China
- National Biosafety Evidence Foundation, Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi’an Jiaotong University, Xi’an 710115, China
| | - Li Zhu
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (H.S.); (L.Z.); (L.S.); (M.L.); (R.W.); (J.Z.)
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Xi’an 710061, China
- National Biosafety Evidence Foundation, Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi’an Jiaotong University, Xi’an 710115, China
| | - Linlan Su
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (H.S.); (L.Z.); (L.S.); (M.L.); (R.W.); (J.Z.)
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Xi’an 710061, China
- National Biosafety Evidence Foundation, Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi’an Jiaotong University, Xi’an 710115, China
| | - Min Li
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (H.S.); (L.Z.); (L.S.); (M.L.); (R.W.); (J.Z.)
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Xi’an 710061, China
- National Biosafety Evidence Foundation, Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi’an Jiaotong University, Xi’an 710115, China
| | - Rui Wang
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (H.S.); (L.Z.); (L.S.); (M.L.); (R.W.); (J.Z.)
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Xi’an 710061, China
- National Biosafety Evidence Foundation, Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi’an Jiaotong University, Xi’an 710115, China
| | - Jie Zhu
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (H.S.); (L.Z.); (L.S.); (M.L.); (R.W.); (J.Z.)
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Xi’an 710061, China
- National Biosafety Evidence Foundation, Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi’an Jiaotong University, Xi’an 710115, China
| | - Yanjiong Chen
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China;
| | - Teng Chen
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (H.S.); (L.Z.); (L.S.); (M.L.); (R.W.); (J.Z.)
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Xi’an 710061, China
- National Biosafety Evidence Foundation, Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi’an Jiaotong University, Xi’an 710115, China
| |
Collapse
|
19
|
Gersamia AG, Pochigaeva KI, Less YE, Akzhigitov RG, Guekht AB, Gulyaeva NV. [Gender characteristics of depressive disorders: clinical, psychological, neurobiological and translational aspects]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:7-16. [PMID: 38529858 DOI: 10.17116/jnevro20241240317] [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] [Indexed: 03/27/2024]
Abstract
Various studies have indicated that the prevalence of depression is almost twice as high among women as among men. A major factor associated with the development of depression and other affective disorders are adverse and psychologically traumatic life events that contribute to changes in the neuroendocrine system, altering the capacity to adapt to stress. These changes are involved in the pathogenesis of mental disorders, along with genetic and other factors, and are to a significant degree regulated by gender dependent mechanisms. While women have a high prevalence of depressive disorders, men show a higher rate of alcohol and substance abuse. These differences in the epidemiology are most likely explained by different predisposition to mental disorders in men and women and a diversity of biological consequences to adverse life events. Taking this into account, there is a need for a critical review of currently used approaches to modeling depressive disorders in preclinical studies, including the use of animals of both sexes. Adaptation of experimental models and protocols taking into account gender characteristics of neuroendocrine changes in response to stress, as well as structural-morphological, electrophysiological, molecular, genetic and epigenetic features, will significantly increase the translational validity of experimental work.
Collapse
Affiliation(s)
- A G Gersamia
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| | - K I Pochigaeva
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| | - Yu E Less
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| | - R G Akzhigitov
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| | - A B Guekht
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - N V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology of RAS, Moscow, Russia
| |
Collapse
|
20
|
Gundacker A, Glat M, Wais J, Stoehrmann P, Pollak A, Pollak DD. Early-life iron deficiency persistently disrupts affective behaviour in mice. Ann Med 2023; 55:1265-1277. [PMID: 37096819 PMCID: PMC10132221 DOI: 10.1080/07853890.2023.2191003] [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: 12/20/2022] [Accepted: 03/09/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND/OBJECTIVE Iron deficiency (ID) is the most common nutrient deficiency, affecting two billion people worldwide, including about 30% of pregnant women. During gestation, the brain is particularly vulnerable to environmental insults, which can irrevocably impair critical developmental processes. Consequently, detrimental consequences of early-life ID for offspring brain structure and function have been described. Although early life ID has been associated with an increased long-term risk for several neuropsychiatric disorders, the effect on depressive disorders has remained unresolved. MATERIALS AND METHODS A mouse model of moderate foetal and neonatal ID was established by keeping pregnant dams on an iron-deficient diet throughout gestation until postnatal day 10. The ensuing significant decrease of iron content in the offspring brain, as well as the impact on maternal behaviour and offspring vocalization was determined in the first postnatal week. The consequences of early-life ID for depression- and anxiety-like behaviour in adulthood were revealed employing dedicated behavioural assays. miRNA sequencing of hippocampal tissue of offspring revealed specific miRNAs signatures accompanying the behavioural deficits of foetal and neonatal ID in the adult brain. RESULTS Mothers receiving iron-deficient food during pregnancy and lactation exhibited significantly less licking and grooming behaviour, while active pup retrieval and pup ultrasonic vocalizations were unaltered. Adult offspring with a history of foetal and neonatal ID showed an increase in depression- and anxiety-like behaviour, paralleled by a deranged miRNA expression profile in the hippocampus, specifically levels of miR200a and miR200b. CONCLUSION ID during the foetal and neonatal periods has life-long consequences for affective behaviour in mice and leaves a specific and persistent mark on the expression of miRNAs in the brain. Foetal and neonatal ID needs to be further considered as risk factor for the development of depression and anxiety disorders later in life.Key MessagesMarginal reduction of gestational alimentary iron intake decreases brain iron content of the juvenile offspring.Early-life ID is associated with increased depression- and anxiety-like behaviour in adulthood.Reduction of maternal alimentary iron intake during pregnancy is reflected in an alteration of miRNA signatures in the adult offspring brain.
Collapse
Affiliation(s)
- Anna Gundacker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Micaela Glat
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jonathan Wais
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Peter Stoehrmann
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Arnold Pollak
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Daniela D. Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
21
|
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: 2.0] [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
|
22
|
Hou KC, Tsai MH, Akbarian S, Huang HS. Mir125b-1 is Not Imprinted in Human Brain and Shows Developmental Expression Changes in Mouse Brain. Neuroscience 2023; 529:99-106. [PMID: 37598835 DOI: 10.1016/j.neuroscience.2023.08.014] [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: 07/19/2023] [Revised: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 08/22/2023]
Abstract
Genomic imprinting is a predominantly brain and placenta-specific epigenetic process that contributes to parent-of-origin-specific gene expression. While microRNAs are highly expressed in the brain, their imprinting status in this tissue remains poorly studied. Previous research demonstrated that Mir125b-2 is imprinted in the human brain and regulates hippocampal circuits and functions in mice. However, the imprinting status of another isoform of miR125b, Mir125b-1, in the human brain, as well as its spatiotemporal expression patterns in mice, have not been elucidated. Here, we show MIR125B1 is not imprinted in the human brain. Moreover, miR-125b-1 was highly expressed in the brains of mice. Furthermore, miR-125b-1 was down-regulated during brain development in mice. Specifically, miR-125b-1 displayed preferential expression in the olfactory bulb, thalamus, and hypothalamus of the mouse brain. Notably, miR-125b-1 was enriched in GABAergic neurons, particularly somatostatin-expressing GABAergic neurons, compared with glutamatergic neurons. Taken together, our findings provide the imprinting status and comprehensive spatiotemporal expression profiling of Mir125b-1 in the brain.
Collapse
Affiliation(s)
- Kuan-Chu Hou
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Meng-Han Tsai
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Schahram Akbarian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Hsien-Sung Huang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.
| |
Collapse
|
23
|
Zhao M, He X, Li T, Shao H, Huo Q, Li Y. Early-Life Factors and Multimorbidity Risk Later in Older Age: Evidence Based on CHARLS. Gerontology 2023; 69:1347-1357. [PMID: 37725916 DOI: 10.1159/000532060] [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: 12/14/2022] [Accepted: 07/17/2023] [Indexed: 09/21/2023] Open
Abstract
INTRODUCTION Early-life factors were reported to exert influence on the health condition of individuals in the long-term. However, limited research explored the connection between early-life factors and multimorbidity in later years. METHODS We utilized the data from the China Health and Retirement Longitudinal Study to assess this possible association in the present cross-sectional study. Multimorbidity was determined based on 14 common chronic diseases included in the study. Logistic regression was employed to examine the link between early-life factors and subsequent multimorbidity. RESULTS Out of 7,578 participants who met the inclusion criteria for analysis, 3,765 (49.68%) were females. The mean age was 68.25 ± 6.70 years. Participants who rated their health during childhood as average (odds ratio [OR] 0.78, 95% confidence interval [CI] 0.63-0.96) or better [OR 0.72, 95% CI 0.57-0.91] were significantly less likely to experience multimorbidity in older life. By contrast, experiencing violence from two of the family members was significantly associated with future multimorbidity (OR [95% CI], 1.29 [1.04-1.60]). A superior family financial situation was also negatively associated with multimorbidity, with average (OR [95% CI], 0.72 [0.63-0.83]) and better off than average (OR [95% CI], 0.76 [0.62-0.93]). DISCUSSION Individuals with poor health status, inferior family socioeconomic status, or experienced violence from family members in childhood were more likely to suffer from multimorbidity in later life. Enhanced social monitoring of potentially adverse conditions in youngsters and targeted interventions could help mitigate the progression of multimorbidity in later life.
Collapse
Affiliation(s)
- Minjun Zhao
- Department of Geriatrics, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China,
| | - Xu He
- Department of Geriatrics, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Tianrong Li
- Department of Geriatrics, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Heng Shao
- Department of Geriatrics, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Qian Huo
- Department of Geriatrics, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Yan Li
- Department of Geriatrics, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
24
|
Zhao J, Zeng X, Liu J, Liu X, Liu Z, Wang B, Chen Z, Dong Y, Guo S, Cui M, Xiao H, Liu X. Marasmius androsaceus mitigates depression-exacerbated intestinal radiation injuries through reprogramming hippocampal miRNA expression. Biomed Pharmacother 2023; 165:115157. [PMID: 37454593 DOI: 10.1016/j.biopha.2023.115157] [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: 05/22/2023] [Revised: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023] Open
Abstract
INTRODUCTION Cancer patients commonly experience high levels of psychological stress, which poses significant risks to their well-being. Radiotherapy is a primary treatment modality for cancer; however, it often leads to intestinal injuries in these patients. Nevertheless, the impact of mental stress on radiotherapy-intertwined complications remains unclear. METHODS To induce intestinal injury, we employed total abdominal irradiation in our experimental model. We conducted high-throughput sequencing to analyze the expression profile of miRNAs in the hippocampus. RESULTS We observed that mice with depression exhibited more severe intestinal injuries following total abdominal irradiation. Remarkably, oral administration of Marasmius androsaceus not only alleviated the depressive phenotype but also mitigated radiation-induced intestinal toxicity. Notably, this radioprotective effect was not observed in mice without depression. Depression disrupted the hippocampal miRNA expression profile in mice subjected to local irradiation of the abdomen, leading to the accumulation of miR-139-5p and miR-184-3p in the hippocampus, serum, and small intestine tissues. However, treatment with Marasmius androsaceus reprogrammed the miRNA expression signature in mice with depression. Furthermore, intravenous injection of antagomirs targeting miR-139-5p and miR-184-3p ameliorated depression, up-regulated Spn expression, reduced radiation enteritis, and improved the integrity of the small intestine in irradiated mice. CONCLUSION Our findings demonstrate the efficacy of Marasmius androsaceus, a small mushroom, in alleviating depression-aggravated intestinal toxicity following radiotherapy by reprogramming hippocampal miRNA expression.
Collapse
Affiliation(s)
- Jiamin Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of MolecularMicrobiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xiaozhou Zeng
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China
| | - Jia Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of MolecularMicrobiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xiaojing Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of MolecularMicrobiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Zhihong Liu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Dushuhu Campus, Soochow University, 199 Ren'ai Road, Suzhou 215123, China
| | - Bin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China
| | - Zhiyuan Chen
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China
| | - Yanxi Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China
| | - Suping Guo
- Shanxi Institute of Medicine and Life Science, 61 Pingyang Road, Taiyuan 030012, China; Shanxi Kangxin Pharmaceutical Co., Ltd, 1 West Ring Road, Luliang 030082, China
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China.
| | - Huiwen Xiao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of MolecularMicrobiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| | - Xingzhong Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of MolecularMicrobiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| |
Collapse
|
25
|
Dantas ESO, Meira KC, Bredemeier J, Amorim KPC. Suicide among women in Brazil: a necessary discussion from a gender perspective. CIENCIA & SAUDE COLETIVA 2023; 28:1469-1477. [PMID: 37194879 DOI: 10.1590/1413-81232023285.16212022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 10/20/2022] [Indexed: 05/18/2023] Open
Abstract
Suicide among women is a matter of public health, and there is a lack of scientific literature on this issue. In this theoretical essay, we sought to discuss suicide among women in Brazil from a gender perspective. For that purpose, we adopted the idea that gender extrapolates the concept of sex, considering that differences between people are produced by culture and arrangements through which society transforms biological sexuality into the realizations of human life. Therefore, this article is organized in a way to indicate some explanatory models of suicide among women, discussing gender inequalities and approaching the matter of intersectionality from a protective view. Moreover, we believe that the theme is extremely complex, considering that stigma still resists, as does prejudice related to this issue. Hence, it is of utmost importance to view the structural questions that refer to suicide in women, such as violence and gender inequalities.
Collapse
Affiliation(s)
- Eder Samuel Oliveira Dantas
- Programa de Pós-Graduação em Saúde Coletiva, Universidade Federal do Rio Grande do Norte (UFRN). Campus Universitário s/n, Lagoa Nova. 59078-970 Natal RN Brasil.
| | | | - Juliana Bredemeier
- Núcleo de Prática Baseada em Evidências, Instituto de Terapia Cognitivo Comportamental. Porto Alegre RS Brasil
| | - Karla Patrícia Cardoso Amorim
- Programa de Pós-Graduação em Saúde Coletiva, Universidade Federal do Rio Grande do Norte (UFRN). Campus Universitário s/n, Lagoa Nova. 59078-970 Natal RN Brasil.
| |
Collapse
|
26
|
van Oers K, van den Heuvel K, Sepers B. The Epigenetics of Animal Personality. Neurosci Biobehav Rev 2023; 150:105194. [PMID: 37094740 DOI: 10.1016/j.neubiorev.2023.105194] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 04/26/2023]
Abstract
Animal personality, consistent individual differences in behaviour, is an important concept for understanding how individuals vary in how they cope with environmental challenges. In order to understand the evolutionary significance of animal personality, it is crucial to understand the underlying regulatory mechanisms. Epigenetic marks such as DNA methylation are hypothesised to play a major role in explaining variation in phenotypic changes in response to environmental alterations. Several characteristics of DNA methylation also align well with the concept of animal personality. In this review paper, we summarise the current literature on the role that molecular epigenetic mechanisms may have in explaining personality variation. We elaborate on the potential for epigenetic mechanisms to explain behavioural variation, behavioural development and temporal consistency in behaviour. We then suggest future routes for this emerging field and point to potential pitfalls that may be encountered. We conclude that a more inclusive approach is needed for studying the epigenetics of animal personality and that epigenetic mechanisms cannot be studied without considering the genetic background.
Collapse
Affiliation(s)
- Kees van Oers
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Wageningen, The Netherlands; Behavioural Ecology Group, Wageningen University & Research (WUR), Wageningen, the Netherlands.
| | - Krista van den Heuvel
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Wageningen, The Netherlands; Behavioural Ecology Group, Wageningen University & Research (WUR), Wageningen, the Netherlands
| | - Bernice Sepers
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Wageningen, The Netherlands; Behavioural Ecology Group, Wageningen University & Research (WUR), Wageningen, the Netherlands
| |
Collapse
|
27
|
Ma X, Li Q, Chen G, Xie J, Wu M, Meng F, Liu J, Liu Y, Zhao D, Wang W, Wang D, Liu C, Dai J, Li C, Cui M. Role of Hippocampal miR-132-3p in Modifying the Function of Protein Phosphatase Mg2+/Mn2+-dependent 1 F in Depression. Neurochem Res 2023:10.1007/s11064-023-03926-8. [PMID: 37036545 DOI: 10.1007/s11064-023-03926-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/15/2023] [Accepted: 03/28/2023] [Indexed: 04/11/2023]
Abstract
Depression is a common, severe, and debilitating psychiatric disorder of unclear etiology. Our previous study has shown that protein phosphatase Mg2+/Mn2+-dependent 1F (PPM1F) in the hippocampal dentate gyrus (DG) displays significant regulatory effects in depression-related behaviors. miR-132-3p plays a potential role in the etiology of depression. This study explored the effect of miR-132-3p on the onset of depression and the possible underlying mechanism for modulating PPM1F expression during the pathology of depression. We found that miR-132-3p levels in the hippocampus of depressed mice subjected to chronic unpredictable stress (CUS) were dramatically reduced, which were correlated with depression-related behaviors. Knockdown of miR-132-3p in hippocampal DG resulted in depression-related phenotypes and increased susceptibility to stress. miR-132-3p overexpression in hippocampal DG alleviated CUS-induced depression-related performance. We then screened out the potential target genes of miR-132-3p, and we found that the expression profiles of sterol regulatory element-binding transcription factor 1 (Srebf1) and forkhead box protein O3a (FOXO3a) were positively correlated with PPM1F under the condition of miR-132-3p knockdown. Finally, as anticipated, we revealed that the activities of Ca2+/calmodulin-dependent protein kinase II (CAMKII) and adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) were reduced, which underlies the target signaling pathway of PPM1F. In conclusion, our study suggests that miR-132-3p was designed to regulate depression-related behaviors by indirectly regulating PPM1F and targeting Srebf1 and FOXO3a, which have been linked to the pathogenesis and treatment of depression.
Collapse
Affiliation(s)
- Xiangxian Ma
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Qiongyu Li
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Guanhong Chen
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- The first clinical medical college, Binzhou Medical University, Yantai, Shandong, China
| | - Junjie Xie
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- The first clinical medical college, Binzhou Medical University, Yantai, Shandong, China
| | - Min Wu
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Fantao Meng
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jing Liu
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yong Liu
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Physiology, Binzhou Medical University, Shandong, China
| | - Di Zhao
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Wentao Wang
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Dan Wang
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Cuilan Liu
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Juanjuan Dai
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chen Li
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China.
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China.
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Minghu Cui
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China.
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China.
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
28
|
Du Y, Wang YL, Chen L, Li QE, Cheng Y. Anti-depressant-like effects of rannasangpei and its active ingredient crocin-1 on chronic unpredictable mild stress mice. Front Pharmacol 2023; 14:1143286. [PMID: 37007014 PMCID: PMC10060548 DOI: 10.3389/fphar.2023.1143286] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Major depressive disorder is one of the most common neuropsychiatric diseases and it is a global public health problem that leads to disabilities. Currently, there is a growing need to explore novel strategy to cure major depressive disorder due to the limitation of available treatments. Rannasangpei (RSNP) is a traditional Tibetan medicine which acts as a therapeutic agent in various acute or chronic diseases, including cardiovascular diseases and neurodegenerative diseases. Crocin-1 a coloring ingredient of saffron which exhibited anti-oxidative and anti-inflammatory properties. Here, we aimed to illustrate whether RSNP and its active ingredient crocin-1 rescue depressive-like phenotypes in chronic unpredictable mild stress (CUMS) induced mouse model of depression. Our results showed that peripheral administration of RSNP or crocin-1 ameliorated the depressive-like behaviors in CUMS-treated mice, as demonstrated by the forced swimming test and tail suspension test. Furthermore, RSNP or crocin-1 treatment reduced oxidative stress in the peripheral blood and hippocampus of the CUMS-treated mice. Additionally, the dysregulated immune system response, as demonstrated by the increased expression of the pro-inflammatory factors (tumor necrosis factor-α and interleukin-6) and the decreased expression of the anti-inflammatory factor-interleukin-10 in the prefrontal cortex and/or hippocampus of CUMS-treated mice, were at least partially restored by RSNP or crocin-1 treatment. RSNP or crocin-1 also restored apoptotic protein marker (Bcl-2 and Bax) levels in the prefrontal cortex and hippocampus of the CUMS-treated mice. Moreover, our data indicated that RSNP or crocin-1 increased astrocyte number and brain-derived neurotrophic factor levels in the hippocampus of CUMS-treated mice after RSNP or crocin-1 administration. Taken together, our study for the first time revealed an anti-depressant effect of RSNP and its active ingredient crocin-1 in a mouse model of depression, with involvement of oxidative stress, inflammatory response and apoptotic pathway.
Collapse
Affiliation(s)
- Yang Du
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Yan-Li Wang
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Lei Chen
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Qi-En Li
- Tibetan Medical College, Qinghai University, Xining, Qinghai, China
- *Correspondence: Qi-En Li, ; Yong Cheng,
| | - Yong Cheng
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
- Institute of National Security, Minzu University of China, Beijing, China
- NHC Key Laboratory of Birth Defect Research, Prevention and Treatment (Hunan Provincial Maternal and Child Healthcare Hospital), Changsha, Hunan, China
- *Correspondence: Qi-En Li, ; Yong Cheng,
| |
Collapse
|
29
|
曾 琪, 廖 婧, 冉 柳, 石 磊, 陈 羽, 张 晨, 向 娇, 洪 素, 况 利. [Sequencing Analysis of miRNAs in Brain-Derived Exosomes of Adolescent Mice With Depression-Like Behaviors]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:316-321. [PMID: 36949692 PMCID: PMC10409158 DOI: 10.12182/20230360205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Indexed: 03/24/2023]
Abstract
Objective To explore the differential expression of microRNAs (miRNAs) in brain-derived exosomes (BDEs) of adolescent mice with depression-like behavior. Methods The experimental group consisted of susceptible adolescent mice exposed to chronic social defeat stress (CSDS), and sucrose preference test (SPT) and open field test (OFT) were performed to evaluate their depression-like behaviors. BDEs were extracted by ultracentrifugation (UC). The morphology, particle size, and surface marker proteins of BDEs were examined by transmission electron microscopy, nano-flow cytometry and Western blot. The expression of miRNA in BDEs was evaluated by high-throughput RNA sequencing. GO enrichment analysis and KEGG pathway enrichment analysis were carried out based on bioinformatics. Results The particle size of BDEs ranged between 50 to 100 nm and they displayed a typical disc-shaped vesicle structure. TSG101 and syntenin, the exosome-positive proteins, were detected. In the BDEs of mice with depression-like behaviors induced by CSDS, 13 miRNAs were significantly upregulated and 4 miRNAs were significantly downregulated. Go and KEGG analysis showed that differentially expressed miRNAs were significantly enriched in PI3K-Akt signaling pathway, axonal guidance, and hypoxic response. Conclusion It was found in this study that exosomal miRNAs in brain tissue might be involved in such biological processes as insulin resistance, neuroplasticity, and hypoxic response, thereby regulating brain functions and causing depression-like behaviors.
Collapse
Affiliation(s)
- 琪 曾
- 重庆医科大学附属大学城医院 精神科 (重庆 401331)Department of Psychiatry, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - 婧 廖
- 重庆医科大学附属大学城医院 精神科 (重庆 401331)Department of Psychiatry, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - 柳毅 冉
- 重庆医科大学附属大学城医院 精神科 (重庆 401331)Department of Psychiatry, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - 磊 石
- 重庆医科大学附属大学城医院 精神科 (重庆 401331)Department of Psychiatry, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - 羽佳 陈
- 重庆医科大学附属大学城医院 精神科 (重庆 401331)Department of Psychiatry, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - 晨钰 张
- 重庆医科大学附属大学城医院 精神科 (重庆 401331)Department of Psychiatry, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - 娇娇 向
- 重庆医科大学附属大学城医院 精神科 (重庆 401331)Department of Psychiatry, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - 素 洪
- 重庆医科大学附属大学城医院 精神科 (重庆 401331)Department of Psychiatry, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - 利 况
- 重庆医科大学附属大学城医院 精神科 (重庆 401331)Department of Psychiatry, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
- 重庆医科大学附属第一医院 精神科 (重庆 400016)Department of Psychiatry,The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
30
|
Silveira PP, Meaney MJ. Examining the biological mechanisms of human mental disorders resulting from gene-environment interdependence using novel functional genomic approaches. Neurobiol Dis 2023; 178:106008. [PMID: 36690304 DOI: 10.1016/j.nbd.2023.106008] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/30/2022] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
We explore how functional genomics approaches that integrate datasets from human and non-human model systems can improve our understanding of the effect of gene-environment interplay on the risk for mental disorders. We start by briefly defining the G-E paradigm and its challenges and then discuss the different levels of regulation of gene expression and the corresponding data existing in humans (genome wide genotyping, transcriptomics, DNA methylation, chromatin modifications, chromosome conformational changes, non-coding RNAs, proteomics and metabolomics), discussing novel approaches to the application of these data in the study of the origins of mental health. Finally, we discuss the multilevel integration of diverse types of data. Advance in the use of functional genomics in the context of a G-E perspective improves the detection of vulnerabilities, informing the development of preventive and therapeutic interventions.
Collapse
Affiliation(s)
- Patrícia Pelufo Silveira
- Department of Psychiatry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
| | - Michael J Meaney
- Department of Psychiatry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada; Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (ASTAR), Singapore; Brain - Body Initiative, Agency for Science, Technology and Research (ASTAR), Singapore.
| |
Collapse
|
31
|
Korobkova L, Morin EL, Aoued H, Sannigrahi S, Garza KM, Siebert ER, Walum H, Cabeen RP, Sanchez MM, Dias BG. RNA in extracellular vesicles during adolescence reveal immune, energetic and microbial imprints of early life adversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.23.529808. [PMID: 36865138 PMCID: PMC9980043 DOI: 10.1101/2023.02.23.529808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Exposure to early life adversity (ELA), including childhood maltreatment, is one of the most significant risk factors for the emergence of neuropsychiatric disorders in adolescence and adulthood. Despite this relationship being well established, the underlying mechanisms remain unclear. One way to achieve this understanding is to identify molecular pathways and processes that are perturbed as a consequence of childhood maltreatment. Ideally, these perturbations would be evident as changes in DNA, RNA or protein profiles in easily accessible biological samples collected in the shadow of childhood maltreatment. In this study, we isolated circulating extracellular vesicles (EVs) from plasma collected from adolescent rhesus macaques that had either experienced nurturing maternal care (CONT) or maternal maltreatment (MALT) in infancy. RNA sequencing of RNA in plasma EVs and gene enrichment analysis revealed that genes related to translation, ATP synthesis, mitochondrial function and immune response were downregulated in MALT samples, while genes involved in ion transport, metabolism and cell differentiation were upregulated. Interestingly, we found that a significant proportion of EV RNA aligned to the microbiome and that MALT altered the diversity of microbiome-associated RNA signatures found in EVs. Part of this altered diversity suggested differences in prevalence of bacterial species in CONT and MALT animals noted in the RNA signatures of the circulating EVs. Our findings provide evidence that immune function, cellular energetics and the microbiome may be important conduits via which infant maltreatment exerts effects on physiology and behavior in adolescence and adulthood. As a corollary, perturbations of RNA profiles related to immune function, cellular energetics and the microbiome may serve as biomarkers of responsiveness to ELA. Our results demonstrate that RNA profiles in EVs can serve as a powerful proxy to identify biological processes that might be perturbed by ELA and that may contribute to the etiology of neuropsychiatric disorders in the aftermath of ELA.
Collapse
|
32
|
Weapons of stress reduction: (R,S)-ketamine and its metabolites as prophylactics for the prevention of stress-induced psychiatric disorders. Neuropharmacology 2023; 224:109345. [PMID: 36427554 DOI: 10.1016/j.neuropharm.2022.109345] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022]
Abstract
Exposure to stress is one of the greatest contributing factors to developing a psychiatric disorder, particularly in susceptible populations. Enhancing resilience to stress could be a powerful intervention to reduce the incidence of psychiatric disease and reveal insight into the pathophysiology of psychiatric disorders. (R,S)-ketamine and its metabolites have recently been shown to exert protective effects when administered before or after a variety of stressors and may be effective, tractable prophylactic compounds against psychiatric disease. Drug dosing, sex, age, and strain in preclinical rodent studies, significantly influence the prophylactic effects of (R,S)-ketamine and related compounds. Due to the broad neurobiological actions of (R,S)-ketamine, a variety of mechanisms have been proposed to contribute to the resilience-enhancing effects of this drug, including altering various transcription factors across the genome, enhancing inhibitory connections from the prefrontal cortex, and increasing synaptic plasticity in the hippocampus. Promisingly, select data have shown that (R,S)-ketamine may be an effective prophylactic against psychiatric disorders, such as postpartum depression (PPD). Overall, this review will highlight a brief history of the prophylactic effects of (R,S)-ketamine, the potential mechanisms underlying its protective actions, and possible future directions for translating prophylactic compounds to the clinic. This article is part of the Special Issue on 'Ketamine and its Metabolites'.
Collapse
|
33
|
Peng S, Zhou Y, Xiong L, Wang Q. Identification of novel targets and pathways to distinguish suicide dependent or independent on depression diagnosis. Sci Rep 2023; 13:2488. [PMID: 36781900 PMCID: PMC9925752 DOI: 10.1038/s41598-023-29101-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
In recent years, postmortem brain studies have revealed that some molecular, cellular, and circuit changes associated with suicide, have an independent or additive effect on depression. The aim of the present study is to identify potential phenotypic, tissue, and sex-specific novel targets and pathways to distinguish depression or suicide from major depressive disorder (MDD) comorbid with suicide. The mRNA expression profiling datasets from two previous independent postmortem brain studies of suicide and depression (GSE102556 and GSE101521) were retrieved from the GEO database. Machine learning analysis was used to differentiate three regrouped gene expression profiles, i.e., MDD with suicide, MDD without suicide, and suicide without depression. Weighted correlation network analysis (WGCNA) was further conducted to identify the key modules and hub genes significantly associated with each of these three sub-phenotypes. TissueEnrich approaches were used to find the essential brain tissues and the difference of tissue enriched genes between depression with or without suicide. Dysregulated gene expression cross two variables, including phenotypes and tissues, were determined by global analysis with Vegan. RRHO analysis was applied to examine the difference in global expression pattern between male and female groups. Using the optimized machine learning model, several ncRNAs and mRNAs with higher AUC and MeanDecreaseGini, including GCNT1P1 and AC092745.1, etc., were identified as potential molecular targets to distinguish suicide with, or without MDD and depression without suicide. WGCNA analysis identified some key modules significantly associated with these three phenotypes, and the gene biological functions of the key modules mainly relate to ncRNA and miRNA processing, as well as oxidoreductase and dehydrogenase activity. Hub genes such as RP11-349A22.5, C20orf196, MAPK8IP3 and RP11-697N18.2 were found in these key modules. TissueEnrich analysis showed that nucleus accumbens and subiculum were significantly changed among the 6 brain regions studied. Global analysis with Vegan and RRHO identified PRS26, ARNT and SYN3 as the most significantly differentially expressed genes across phenotype and tissues, and there was little overlap between the male and female groups. In this study, we have identified novel gene targets, as well as annotated functions of co-expression patterns and hub genes that are significantly distinctive between depression with suicide, depression without suicide, and suicide without depression. Moreover, global analysis across three phenotypes and tissues confirmed the evidence of sex difference in mood disorders.
Collapse
Affiliation(s)
- Siqi Peng
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yalan Zhou
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lan Xiong
- Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada.
| | - Qingzhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
34
|
D'Addario SL, Municchi D, Mancini C, Ielpo D, Babicola L, Di Segni M, Iacono LL, Ferlazzo F, Cifani C, Andolina D, Ventura R. The long-lasting effects of early life adversities are sex dependent: The signature of miR-34a. J Affect Disord 2023; 322:277-288. [PMID: 36414112 DOI: 10.1016/j.jad.2022.11.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 10/26/2022] [Accepted: 11/11/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Exposure to early life adversities (ELA) can influence a plethora of biological mechanisms leading to stress-related disorders later in life through epigenetic mechanisms, such as microRNAs (miRs). MiR-34 is a critical modulator of stress response and stress-induced pathologies and a link between ELA and miR-34a has been reported. METHODS Here using our well-established model of ELA (Repeated Cross Fostering) we investigate the behavioral long-term effects of ELA in male and female mice. We also assess basal and ELA-induced miR-34a expression in adult mice and investigate whether ELA affects the later miR-34a response to adult acute stress exposure across brain areas (medial preFrontal Cortex, Dorsal Raphe Nuclei) and peripheral organs (heart, plasma) in animals from both sexes. Finally, based on our previous data demonstrating the critical role of Dorsal Raphe Nuclei miR-34a expression in serotonin (5-HT) transmission, we also investigated prefrontal-accumbal 5-HT outflow induced by acute stress exposure in ELA and Control females by in vivo intracerebral microdialysis. RESULTS ELA not just induces a depressive-like state as well as enduring changes in miR-34a expression, but also alters miR-34a expression in response to adult acute stress exclusively in females. Finally, altered DRN miR-34a expression is associated with prefrontal-accumbal 5-HT release under acute stress exposure in females. LIMITATIONS Translational study on humans is necessary to verify the results obtained in our animal models of ELA-induced depression. CONCLUSIONS This is the first evidence showing long-lasting sex related effects of ELA on brain and peripheral miR-34a expression levels in an animal model of depression-like phenotype.
Collapse
Affiliation(s)
- Sebastian Luca D'Addario
- IRCCS Fondazione Santa Lucia, Roma, Italy; Dept. of Psychology and Center "Daniel Bovet", Sapienza University, Rome 00184, Italy.
| | - Diana Municchi
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, Rome 00184, Italy.
| | - Camilla Mancini
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy.
| | - Donald Ielpo
- IRCCS Fondazione Santa Lucia, Roma, Italy; Dept. of Psychology and Center "Daniel Bovet", Sapienza University, Rome 00184, Italy.
| | - Lucy Babicola
- IRCCS Fondazione Santa Lucia, Roma, Italy; Dept. of Psychology and Center "Daniel Bovet", Sapienza University, Rome 00184, Italy.
| | | | - Luisa Lo Iacono
- IRCCS Fondazione Santa Lucia, Roma, Italy; Dept. of Psychology and Center "Daniel Bovet", Sapienza University, Rome 00184, Italy.
| | - Fabio Ferlazzo
- Dept. of Psychology and Center "Daniel Bovet", Sapienza University, Rome 00184, Italy.
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy.
| | - Diego Andolina
- IRCCS Fondazione Santa Lucia, Roma, Italy; Dept. of Psychology and Center "Daniel Bovet", Sapienza University, Rome 00184, Italy.
| | - Rossella Ventura
- IRCCS Fondazione Santa Lucia, Roma, Italy; Dept. of Psychology and Center "Daniel Bovet", Sapienza University, Rome 00184, Italy.
| |
Collapse
|
35
|
Chen Q, Liu X, Xu R, Wang X, Zhou D, Tang Y. Circulating exosomal microRNAs in nonsuicidal self-injury. Asian J Psychiatr 2023; 80:103310. [PMID: 36470192 DOI: 10.1016/j.ajp.2022.103310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/29/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Qianqian Chen
- Department of Psychosomatic, Taizhou Second People's Hospital, Taizhou, Zhejiang 317200, China
| | - Xilaoli Liu
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang 315201, China
| | - Ruizhi Xu
- Department of Psychosomatic, Taizhou Second People's Hospital, Taizhou, Zhejiang 317200, China
| | - Xiaojing Wang
- Department of Psychosomatic, Taizhou Second People's Hospital, Taizhou, Zhejiang 317200, China
| | - Dongsheng Zhou
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang 315201, China.
| | - Yiping Tang
- Department of Psychosomatic, Taizhou Second People's Hospital, Taizhou, Zhejiang 317200, China.
| |
Collapse
|
36
|
Cannabidiol Modulates Alterations in PFC microRNAs in a Rat Model of Depression. Int J Mol Sci 2023; 24:ijms24032052. [PMID: 36768376 PMCID: PMC9953518 DOI: 10.3390/ijms24032052] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/11/2023] [Accepted: 01/14/2023] [Indexed: 01/21/2023] Open
Abstract
Cannabidiol (CBD) is a potential antidepressant agent. We examined the association between the antidepressant effects of CBD and alterations in brain microRNAs in the unpredictable chronic mild stress (UCMS) model for depression. UCMS male rats were injected with vehicle or CBD (10 mg/kg) and tested for immobility time in the forced swim test. Alterations in miRNAs (miR16, miR124, miR135a) and genes that encode for the 5HT1a receptor, the serotonergic transporter SERT, β-catenin, and CB1 were examined. UCMS increased immobility time in a forced swim test (i.e., depressive-like behavior) and altered the expression of miRNAs and mRNA in the ventromedial prefrontal cortex (vmPFC), raphe nucleus, and nucleus accumbens. Importantly, CBD restored UCMS-induced upregulation in miR-16 and miR-135 in the vmPFC as well as the increase in immobility time. CBD also restored the UCMS-induced decrease in htr1a, the gene that encodes for the serotonergic 5HT1a receptor; using a pharmacological approach, we found that the 5HT1a receptor antagonist WAY100135 blocked the antidepressant-like effect of CBD on immobility time. Our findings suggest that the antidepressant effects of CBD in a rat model for depression are associated with alterations in miR-16 and miR-135 in the vmPFC and are mediated by the 5HT1a receptor.
Collapse
|
37
|
Han K, Ji L, Wang C, Shao Y, Chen C, Liu L, Feng M, Yang F, Wu X, Li X, Xie Q, He L, Shi Y, He G, Dong Z, Yu T. The host genetics affects gut microbiome diversity in Chinese depressed patients. Front Genet 2023; 13:976814. [PMID: 36699448 PMCID: PMC9868868 DOI: 10.3389/fgene.2022.976814] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
The gut microbiome and host genetics are both associated with major depressive disorder (MDD); however, the molecular mechanisms among the associations are poorly understood, especially in the Asian, Chinese group. Our study applied linear discriminant analysis (LDA) effect size (LEfSe) and genome-wide association analysis in the cohort with both gut sequencing data and genomics data. We reported the different gut microbiota characteristics between MDD and control groups in the Chinese group and further constructed the association between host genetics and the gut microbiome. Actinobacteria and Pseudomonades were found more in the MDD group. We found significant differences in the ACE and Chao indexes of alpha diversity while no discrepancy in beta diversity. We found three associations between host genetics with microbiome features: beta diversity and rs6108 (p = 8.65 × 10-9), Actinobacteria and rs77379751 (p = 8.56 × 10-9), and PWY-5913 and rs1775633082 (p = 4.54 × 10-8). A species of the Romboutsia genus was co-associated with the species of Ruminococcus gnavus in an internetwork through four genes: METTL8, ITGB2, OTULIN, and PROSER3, with a strict threshold (p < 5 × 10-4). Furthermore, our findings suggested that the gut microbiome diversity might affect microRNA expression in the brain and influenced SERPINA5 and other spatially close genes afterward. These findings suggest new linkages between depression and gut microbiome in Asian, Chinese people, which might be mediated by genes and microRNA regulation in space distance.
Collapse
Affiliation(s)
- Ke Han
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Ji
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Chenliu Wang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Shao
- Asbios (Tianjin) Biotechnology Co., Ltd., Tianjin, China
| | - Changfeng Chen
- School of Mental Health, Jining Medical University, Jining, China
| | - Liangjie Liu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Mofan Feng
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Fengping Yang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Xi Wu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Xingwang Li
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Qinglian Xie
- Out-patient Department of West China Hospital, Sichuan University, Chengdu, China
| | - Lin He
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Shi
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Guang He
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Zaiquan Dong
- Mental Health Center of West China Hospital, Sichuan University, Chengdu, China
| | - Tao Yu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Women and Children’s Health, Shanghai, China
| |
Collapse
|
38
|
Cabana-Domínguez J, Antón-Galindo E, Fernàndez-Castillo N, Singgih EL, O'Leary A, Norton WH, Strekalova T, Schenck A, Reif A, Lesch KP, Slattery D, Cormand B. The translational genetics of ADHD and related phenotypes in model organisms. Neurosci Biobehav Rev 2023; 144:104949. [PMID: 36368527 DOI: 10.1016/j.neubiorev.2022.104949] [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: 07/01/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a highly prevalent neurodevelopmental disorder resulting from the interaction between genetic and environmental risk factors. It is well known that ADHD co-occurs frequently with other psychiatric disorders due, in part, to shared genetics factors. Although many studies have contributed to delineate the genetic landscape of psychiatric disorders, their specific molecular underpinnings are still not fully understood. The use of animal models can help us to understand the role of specific genes and environmental stimuli-induced epigenetic modifications in the pathogenesis of ADHD and its comorbidities. The aim of this review is to provide an overview on the functional work performed in rodents, zebrafish and fruit fly and highlight the generated insights into the biology of ADHD, with a special focus on genetics and epigenetics. We also describe the behavioral tests that are available to study ADHD-relevant phenotypes and comorbid traits in these models. Furthermore, we have searched for new models to study ADHD and its comorbidities, which can be useful to test potential pharmacological treatments.
Collapse
Affiliation(s)
- Judit Cabana-Domínguez
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain.
| | - Ester Antón-Galindo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain
| | - Noèlia Fernàndez-Castillo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain
| | - Euginia L Singgih
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Aet O'Leary
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany; Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Tartu, Estonia
| | - William Hg Norton
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Tatyana Strekalova
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany, and Department of Neuropsychology and Psychiatry, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany, and Department of Neuropsychology and Psychiatry, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - David Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain.
| |
Collapse
|
39
|
Li W, Li X, Li Y, Chen Y, Zhu L, Guo R. Diagnostic value of MicroRNAs for depression: A systematic review and meta-analysis. J Psychiatr Res 2023; 157:132-140. [PMID: 36463628 DOI: 10.1016/j.jpsychires.2022.11.028] [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: 08/04/2022] [Revised: 11/10/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Currently, depression is diagnosed on the basis of neuropsychological examinations and clinical symptoms, and there is no objective diagnostic method. Several studies have explored the application of microRNAs as potential biomarkers diagnostic for depression. This study aims to determine the diagnostic value of microRNAs for depression. METHODS PubMed, Embase, the Cochrane Library, the Web of Science, Wanfang Database, SINOMED, China Science and Technology Journal Databaseand China National Knowledge Infrastructure were searched up to 11 January 2022. Stata (version 16.0) and RevMan (version 5.3) software were used for meta-analysis. The pooled sensitivity, pooled specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio (DOR) were calculated; the summary receiver operating characteristic (SROC) curve was plotted, and the area under the curve (AUC) was calculated. Moreover, meta-regression analyses were performed to determine the source of heterogeneity. Deeks' funnel plot test was used to assess publication bias. RESULTS In total, 677 patients were enrolled, including 364 patients with depression and 313 healthy controls. Meta-analysis results showed that the pooled sensitivity, specificity, and DOR of microRNAs for the diagnosis of depression were 0.82 [95% confidence intervals(CI): 0.76, 0.87], 0.70 (95% CI: 0.62, 0.77), and 11 (95% CI: 6, 20), respectively, and the AUC of the SROC was 0.84 (95% CI: 0.80, 0.87). CONCLUSIONS MicroRNAs have high sensitivity and specificity in diagnosing depression and are potential diagnostic biomarkers for depression. REGISTRATION NUMBER PROSPERO CRD42022303616.
Collapse
Affiliation(s)
- Wenhui Li
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xingxing Li
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yannan Li
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yi Chen
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Lingqun Zhu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing Key Laboratory of Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Rongjuan Guo
- Department of Neurology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China.
| |
Collapse
|
40
|
Dissecting early life stress-induced adolescent depression through epigenomic approach. Mol Psychiatry 2023; 28:141-153. [PMID: 36517640 PMCID: PMC9812796 DOI: 10.1038/s41380-022-01907-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022]
Abstract
Early life stress (ELS), such as abuse and neglect during childhood, can lead to psychiatric disorders in later life. Previous studies have suggested that ELS can cause profound changes in gene expression through epigenetic mechanisms, which can lead to psychiatric disorders in adulthood; however, studies on epigenetic modifications associated with ELS and psychiatric disorders in adolescents are limited. Moreover, how these epigenetic modifications can lead to psychiatric disorders in adolescents is not fully understood. Commonly, DNA methylation, histone modification, and the regulation of noncoding RNAs have been attributed to the reprogramming of epigenetic profiling associated with ELS. Although only a few studies have attempted to examine epigenetic modifications in adolescents with ELS, existing evidence suggests that there are commonalities and differences in epigenetic profiling between adolescents and adults. In addition, epigenetic modifications are sex-dependent and are influenced by the type of ELS. In this review, we have critically evaluated the current evidence on epigenetic modifications in adolescents with ELS, particularly DNA methylation and the expression of microRNAs in both preclinical models and humans. We have also clarified the impact of ELS on psychiatric disorders in adolescents to predict the development of neuropsychiatric disorders and to prevent and recover these disorders through personalized medicine.
Collapse
|
41
|
Tavares GA, Torres A, Le Drean G, Queignec M, Castellano B, Tesson L, Remy S, Anegon I, Pitard B, Kaeffer B. Oral Delivery of miR-320-3p with Lipidic Aminoglycoside Derivatives at Mid-Lactation Alters miR-320-3p Endogenous Levels in the Gut and Brain of Adult Rats According to Early or Regular Weaning. Int J Mol Sci 2022; 24:ijms24010191. [PMID: 36613633 PMCID: PMC9820440 DOI: 10.3390/ijms24010191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/14/2022] [Accepted: 12/17/2022] [Indexed: 12/25/2022] Open
Abstract
To investigate if the artificial delivery of microRNAs naturally present in the breastmilk can impact the gut and brain of young rats according to weaning. Animals from a new transgenic rat line expressing the green-fluorescent protein in the endocrine lineage (cholecystokinin expressing cells) received a single oral bolus of miR-320-3p or miR-375-3p embedded in DiOleyl-Succinyl-Paromomycin (DOSP) on D-12. The pups were weaned early (D-15), or regularly (D-30). The expression of relevant miRNA, mRNAs, chromatin complexes, and duodenal cell density were assessed at 8 h post-inoculation and on D-45. The miR-320-3p/DOSP induced immediate effects on H3K4me3 chromatin complexes with polr3d promoter (p < 0.05). On regular weaning, on D-45, miR-320-3p and 375-3p were found to be downregulated in the stomach and upregulated in the hypothalamus (p < 0.001), whereas miR-320-3p was upregulated in the duodenum. After early weaning, miR-320-3p and miR-375-3p were downregulated in the stomach and the duodenum, but upregulated in the hypothalamus and the hippocampus. Combination of miR-320-3p/DOSP with early weaning enhanced miR-320-3p and chromogranin A expression in the duodenum. In the female brain stem, miR-320-3p, miR-504, and miR-16-5p levels were all upregulated. Investigating the oral miRNA-320-3p loads in the duodenal cell lineage paved the way for designing new therapeutics to avoid unexpected long-term impacts on the brain.
Collapse
Affiliation(s)
- Gabriel Araujo Tavares
- Nantes Université, INRAE, UMR 1280, PhAN, F-44000 Nantes, France
- Laboratory of Neuroplasticity and Behavior, Graduate Program of Nutrition, Federal University of Pernambuco, Recife 56070-901, Brazil
| | - Amada Torres
- Nantes Université, INRAE, UMR 1280, PhAN, F-44000 Nantes, France
| | - Gwenola Le Drean
- Nantes Université, INRAE, UMR 1280, PhAN, F-44000 Nantes, France
| | - Maïwenn Queignec
- Nantes Université, INRAE, UMR 1280, PhAN, F-44000 Nantes, France
| | | | - Laurent Tesson
- Platform Rat Transgenesis ImmunoPhenomic, INSERM UMR 1064-CRTI, SFR François Bonamy, CNRS UMS3556, F-44093 Nantes, France
| | - Séverine Remy
- Platform Rat Transgenesis ImmunoPhenomic, INSERM UMR 1064-CRTI, SFR François Bonamy, CNRS UMS3556, F-44093 Nantes, France
| | - Ignacio Anegon
- Platform Rat Transgenesis ImmunoPhenomic, INSERM UMR 1064-CRTI, SFR François Bonamy, CNRS UMS3556, F-44093 Nantes, France
| | - Bruno Pitard
- Nantes Université, Univ Angers, INSERM, CNRS, Immunology and New Concepts in Immunotherapy, INCIT UMR1302/EMR6001, F-44000 Nantes, France
| | - Bertrand Kaeffer
- Nantes Université, INRAE, UMR 1280, PhAN, F-44000 Nantes, France
- Correspondence:
| |
Collapse
|
42
|
FAAH Inhibition Restores Early Life Stress-Induced Alterations in PFC microRNAs Associated with Depressive-Like Behavior in Male and Female Rats. Int J Mol Sci 2022; 23:ijms232416101. [PMID: 36555739 PMCID: PMC9782513 DOI: 10.3390/ijms232416101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Early life stress (ELS) increases predisposition to depression. We compared the effects of treatment with the fatty acid amide hydrolase (FAAH) inhibitor URB597, and the selective serotonin reuptake inhibitor paroxetine, on ELS-induced depressive-like behavior and the expression of microRNAs (miRs) associated with depression in the medial prefrontal cortex (mPFC), hippocampal CA1 area, lateral habenula and dorsal raphe in rats. We also examined the mRNA expression of serotonergic (htr1a and slc6a4) and endocannabinoid (cnr1, cnr2 and faah) targets in the mPFC following ELS and pharmacological treatment. Adult males and females exposed to the 'Limited Bedding and Nesting' ELS paradigm demonstrated a depressive-like phenotype and late-adolescence URB597 treatment, but not paroxetine, reversed this phenotype. In the mPFC, ELS downregulated miR-16 in males and miR-135a in females and URB597 treatment restored this effect. In ELS females, the increase in cnr2 and decrease in faah mRNAs in the mPFC were reversed by URB597 treatment. We show for the first time that URB597 reversed ELS-induced mPFC downregulation in specific miRs and stress-related behaviors, suggesting a novel mechanism for the beneficial effects of FAAH inhibition. The differential effects of ELS and URB597 on males and females highlight the importance of developing sex-specific treatment approaches.
Collapse
|
43
|
Ma L, Wang L, Chang L, Shan J, Qu Y, Wang X, Fujita Y, Hashimoto K. A role of microRNA-149 in the prefrontal cortex for prophylactic actions of (R)-ketamine in inflammation model. Neuropharmacology 2022; 219:109250. [PMID: 36088985 DOI: 10.1016/j.neuropharm.2022.109250] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/02/2022] [Accepted: 09/04/2022] [Indexed: 12/12/2022]
Abstract
MicroRNAs (or miRNAs) are short, regulatory RNAs that act as post-transcriptional repressors of gene expression. Recently, we reported that the nuclear factor of activated T cells 4 (NFATc4) signaling might contribute to sustained prophylactic effects of new antidepressant (R)-ketamine in lipopolysaccharide (LPS)-treated inflammation model of depression. In this study, we examined the role of miRNAs (miR-149 and miR-7688-5p) which can regulate NFATc4 in the prefrontal cortex (PFC) of male mice after administration of LPS (1.0 mg/kg). There was a positive correlation between the expression of Nfatc4 and the expression of miR-149 in the PFC. There was also a negative correlation between gene expression of Nfatc4 and gene expression of miR-7688-5p in the PFC. Gut microbiota analysis showed that pretreatment with (R)-ketamine (10 mg/kg) could restore altered composition of gut microbiota in LPS-treated mice. A network analysis showed that gut microbiota may regulate gene expression of Nfatc4 and miR-149 (or miR-7688-5p) in the PFC. Finally, inhibition of miR-149 by antagomiR-149 blocked LPS-induced depression-like behavior by attenuating LPS-induced expression of NFATc4 in the PFC. These findings suggest that the regulation of NFATc4 signaling by miR-149 might play a role in persistent prophylactic effects of (R)-ketamine, and that gut microbiota may regulate the gene expression of miRNAs in the PFC through gut-microbiota-brain axis.
Collapse
Affiliation(s)
- Li Ma
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan; Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Long Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Lijia Chang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Jiajing Shan
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Youge Qu
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Xingming Wang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Yuko Fujita
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| |
Collapse
|
44
|
Therapeutic Implications of microRNAs in Depressive Disorders: A Review. Int J Mol Sci 2022; 23:ijms232113530. [PMID: 36362315 PMCID: PMC9658840 DOI: 10.3390/ijms232113530] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs are hidden players in complex psychophysical phenomena such as depression and anxiety related disorders though the activation and deactivation of multiple proteins in signaling cascades. Depression is classified as a mood disorder and described as feelings of sadness, loss, or anger that interfere with a person’s everyday activities. In this review, we have focused on exploration of the significant role of miRNAs in depression by affecting associated target proteins (cellular and synaptic) and their signaling pathways which can be controlled by the attachment of miRNAs at transcriptional and translational levels. Moreover, miRNAs have potential role as biomarkers and may help to cure depression through involvement and interactions with multiple pharmacological and physiological therapies. Taken together, miRNAs might be considered as promising novel therapy targets themselves and may interfere with currently available antidepressant treatments.
Collapse
|
45
|
Ran LY, Kong YT, Xiang JJ, Zeng Q, Zhang CY, Shi L, Qiu HT, Liu C, Wu LL, Li YL, Chen JM, Ai M, Wang W, Kuang L. Serum extracellular vesicle microRNA dysregulation and childhood trauma in adolescents with major depressive disorder. Bosn J Basic Med Sci 2022; 22:959-971. [PMID: 35659238 PMCID: PMC9589301 DOI: 10.17305/bjbms.2022.7110] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/23/2022] [Indexed: 07/20/2023] Open
Abstract
Major depressive disorder (MDD) seriously endangers adolescent mental and physical health. Extracellular vesicles (EVs) are mediators of cellular communication and are involved in many physiological brain processes. Although EV miRNAshave been implicated in adults with major psychiatric disorders, investigation into their effects in adolescent MDDremains scarce. In discovery set, we conducted a genome-wide miRNA sequencing of serum EVs from 9 untreated adolescents with MDD and 8 matched healthy controls (HCs), identifying 32 differentially expressed miRNAs (18 upregulated and 14 downregulated). In the validation set, 8 differentially expressed and highly enriched miRNAs were verified in independent samples using RT-PCR, with 4 (miR-450a-2-3p, miR-3691-5p, miR-556-3p, and miR-2115-3p) of the 8 miRNAs found to be significantly elevated in 34 untreated adolescents with MDD compared with 38 HCs and consistent with the sequencing results. After the Bonferroni correction, we found that three miRNAs (miR-450a-2-3p, miR-556-3p, and miR-2115-3p) were still significantly different. Among them, miR-450a-2-3p showed the most markeddifferential expression and was able to diagnose disease with 67.6% sensitivity and 84.2% specificity. Furthermore, miR-450a-2-3p partially mediated the associations between total childhood trauma, emotional abuse, and physical neglect and adolescent MDD. We also found that the combination of miR-450a-2-3p and emotional abuse could effectively diagnose MDD in adolescents with 82.4% sensitivity and 81.6% specificity. Our data demonstrate the association of serum EV miRNA dysregulation with MDD pathophysiology and, furthermore, show that miRNAs may mediate the relationship between early stress and MDD susceptibility. We also provide a valid integrated model for the diagnosis of adolescent MDD.
Collapse
Affiliation(s)
- Liu-Yi Ran
- Mental Health Center, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Yi-Ting Kong
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiao-Jiao Xiang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qi Zeng
- Mental Health Center, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Chen-Yu Zhang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Shi
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hai-Tang Qiu
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chuan Liu
- Mental Health Center, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Lin-Li Wu
- Mental Health Center, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Ya-Lan Li
- Mental Health Center, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Jian-Mei Chen
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Ai
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wo Wang
- Mental Health Center, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Li Kuang
- Mental Health Center, University-Town Hospital of Chongqing Medical University, Chongqing, China
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
46
|
Ma L, Wang L, Chang L, Shan J, Qu Y, Wang X, Wan X, Fujita Y, Hashimoto K. A key role of miR-132-5p in the prefrontal cortex for persistent prophylactic actions of (R)-ketamine in mice. Transl Psychiatry 2022; 12:417. [PMID: 36171191 PMCID: PMC9519951 DOI: 10.1038/s41398-022-02192-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/08/2022] [Accepted: 09/16/2022] [Indexed: 11/09/2022] Open
Abstract
(R,S)-ketamine is known to elicit persistent prophylactic effects in rodent models of depression. However, the precise molecular mechanisms underlying its action remain elusive. Using RNA-sequencing analysis, we searched for novel molecular target(s) that contribute to the prophylactic effects of (R)-ketamine, a more potent enantiomer of (R,S)-ketamine in chronic restraint stress (CRS) model. Pretreatment with (R)-ketamine (10 mg/kg, 1 day before CRS) significantly ameliorated body weight loss, increased immobility time of forced swimming test, and decreased sucrose preference of sucrose preference test in CRS-exposed mice. RNA-sequencing analysis of prefrontal cortex (PFC) revealed that several miRNAs such as miR-132-5p might contribute to sustained prophylactic effects of (R)-ketamine. Methyl CpG binding protein 2 (MeCP2) is known to regulate brain-derived neurotrophic factor (BDNF) expression. Quantitative RT-PCR confirmed that (R)-ketamine significantly attenuated altered expression of miR-132-5p and its regulated genes (Bdnf, Mecp2, Tgfb1, Tgfbr2) in the PFC of CRS-exposed mice. Furthermore, (R)-ketamine significantly attenuated altered expression of BDNF, MeCP2, TGF-β1 (transforming growth factor β1), and synaptic proteins (PSD-95, and GluA1) in the PFC of CRS-exposed mice. Administration of agomiR-132-5p decreased the expression of Bdnf and Tgfb1 in the PFC, resulting in depression-like behaviors. In contrast, administration of antagomiR-132-5p blocked the increased expression of miR-132-5p and decreased expression of Bdnf in the PFC of CRS-exposed mice, resulting in antidepressant-like effects. In conclusion, our data show a novel role of miR-132-5p in the PFC underlying depression-like phenotypes in CRS model and the sustained prophylactic effects of (R)-ketamine.
Collapse
Affiliation(s)
- Li Ma
- grid.411500.1Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan ,grid.412632.00000 0004 1758 2270Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province China
| | - Long Wang
- grid.412632.00000 0004 1758 2270Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province China
| | - Lijia Chang
- grid.411500.1Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Jiajing Shan
- grid.411500.1Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Youge Qu
- grid.411500.1Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Xingming Wang
- grid.411500.1Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Xiayun Wan
- grid.411500.1Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Yuko Fujita
- grid.411500.1Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan.
| |
Collapse
|
47
|
Šalamon Arčan I, Kouter K, Videtič Paska A. Depressive disorder and antidepressants from an epigenetic point of view. World J Psychiatry 2022; 12:1150-1168. [PMID: 36186508 PMCID: PMC9521527 DOI: 10.5498/wjp.v12.i9.1150] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/27/2022] [Accepted: 08/05/2022] [Indexed: 02/05/2023] Open
Abstract
Depressive disorder is a complex, heterogeneous disease that affects approximately 280 million people worldwide. Environmental, genetic, and neurobiological factors contribute to the depressive state. Since the nervous system is susceptible to shifts in activity of epigenetic modifiers, these allow for significant plasticity and response to rapid changes in the environment. Among the most studied epigenetic modifications in depressive disorder is DNA methylation, with findings centered on the brain-derived neurotrophic factor gene, the glucocorticoid receptor gene, and the serotonin transporter gene. In order to identify biomarkers that would be useful in clinical settings, for diagnosis and for treatment response, further research on antidepressants and alterations they cause in the epigenetic landscape throughout the genome is needed. Studies on cornerstone antidepressants, such as selective serotonin reuptake inhibitors, selective serotonin and norepinephrine reuptake inhibitors, norepinephrine, and dopamine reuptake inhibitors and their effects on depressive disorder are available, but systematic conclusions on their effects are still hard to draw due to the highly heterogeneous nature of the studies. In addition, two novel drugs, ketamine and esketamine, are being investigated particularly in association with treatment of resistant depression, which is one of the hot topics of contemporary research and the field of precision psychiatry.
Collapse
Affiliation(s)
- Iris Šalamon Arčan
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana SI-1000, Slovenia
| | - Katarina Kouter
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana SI-1000, Slovenia
| | - Alja Videtič Paska
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana SI-1000, Slovenia
| |
Collapse
|
48
|
Lahiri DK, Maloney B, Song W, Sokol DK. Crossing the "Birth Border" for Epigenetic Effects. Biol Psychiatry 2022; 92:e21-e23. [PMID: 35248366 PMCID: PMC9514510 DOI: 10.1016/j.biopsych.2021.10.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/17/2021] [Indexed: 11/18/2022]
Affiliation(s)
- Debomoy K Lahiri
- Department of Psychiatry, Indiana Alzheimer's Disease Research Center, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana.
| | - Bryan Maloney
- Department of Psychiatry, Indiana Alzheimer's Disease Research Center, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Weihong Song
- Institute of Aging, Wenzhou Medical University, Wenzhou, China; Department of Psychiatry, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Deborah K Sokol
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
49
|
Vulnerabilidade à inatividade física: validação de conteúdo dos marcadores para adultos. ACTA PAUL ENFERM 2022. [DOI: 10.37689/acta-ape/2022ar020766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
50
|
Cell-type-specific epigenetic effects of early life stress on the brain. Transl Psychiatry 2022; 12:326. [PMID: 35948532 PMCID: PMC9365848 DOI: 10.1038/s41398-022-02076-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 02/08/2023] Open
Abstract
Early life stress (ELS) induces long-term phenotypic adaptations that contribute to increased vulnerability to a host of neuropsychiatric disorders. Epigenetic mechanisms, including DNA methylation, histone modifications and non-coding RNA, are a proposed link between environmental stressors, alterations in gene expression, and phenotypes. Epigenetic modifications play a primary role in shaping functional differences between cell types and can be modified by environmental perturbations, especially in early development. Together with contributions from genetic variation, epigenetic mechanisms orchestrate patterns of gene expression within specific cell types that contribute to phenotypic variation between individuals. To date, many studies have provided insights into epigenetic changes resulting from ELS. However, most of these studies have examined heterogenous brain tissue, despite evidence of cell-type-specific epigenetic modifications in phenotypes associated with ELS. In this review, we focus on rodent and human studies that have examined epigenetic modifications induced by ELS in select cell types isolated from the brain or associated with genes that have cell-type-restricted expression in neurons, microglia, astrocytes, and oligodendrocytes. Although significant challenges remain, future studies using these approaches can enable important mechanistic insight into the role of epigenetic variation in the effects of ELS on brain function.
Collapse
|