1
|
Chen H, Ouyang W, Cui X, Ma X, Hu S, Qing W, Tong J. miR-124 mediates the effects of gut microbial dysbiosis on brain function in chronic stressed mice. Behav Brain Res 2025; 476:115262. [PMID: 39306097 DOI: 10.1016/j.bbr.2024.115262] [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: 02/19/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024]
Abstract
The gut microbiota plays a key role in the brain function impairment caused by chronic stress, yet its exact mechanism remains unclear. Many studies have revealed the important role of miR-124 in the central nervous system. Meanwhile, previous studies have indicated that miR-124 may be regulated by chronic stress and gut microbiota. Here, we aimed to explore whether miR-124 serves as a mediator for the impacts of gut microbial dysbiosis on brain function in mice subjected to chronic stress. Repeated daily restraint stress for 4 weeks was used to induce chronic stress in mice. Chronic stress resulted in gut microbial dysbiosis, abnormal behaviors, and a decrease in hippocampal miR-124 levels. Treatment with different probiotic mixtures significantly alleviated the effects of chronic stress on hippocampal miR-124 levels and mouse behaviors. Suppression of hippocampal miR-124 expression reversed the beneficial effects of probiotics on cognitive function, neurogenesis, and related molecular markers in chronically stressed mice. Bioinformatics analysis and qPCR suggested that Ptpn11 might be a target gene for miR-124 in mediating the effects of gut microbial dysbiosis on brain function in these mice. These findings suggest that miR-124 is a pivotal regulator that mediates the detrimental effects of gut microbial dysbiosis on brain function and the subsequent cognitive impairment during chronic stress.
Collapse
Affiliation(s)
- Hui Chen
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China; Department of Laboratory Medicine, Third Xiangya Hospital, Central South University, Changsha 410013, China; Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Wen Ouyang
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China; Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xiaoyu Cui
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China; Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xin Ma
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China; Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Shanshan Hu
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China; Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Wenxiang Qing
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China; Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Jianbin Tong
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China; Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha 410013, China; Center for Experimental Medicine, Third Xiangya Hospital, Central South University, Changsha 410013, China; Brain Research Center, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
2
|
Guan W, Wu XY, Jin X, Sheng XM, Fan Y. miR-204-5p Plays a Critical Role in the Pathogenesis of Depression and Anti-depression Action of Venlafaxine in the Hippocampus of Mice. Curr Med Chem 2024; 31:3412-3425. [PMID: 37357509 DOI: 10.2174/0929867330666230623163315] [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: 12/31/2022] [Revised: 04/04/2023] [Accepted: 04/10/2023] [Indexed: 06/27/2023]
Abstract
BACKGROUND Venlafaxine has been demonstrated to treat diseases such as social anxiety disorder and depression. Most of antidepressants including venlafaxine have a certain effect, but significant side effects. Therefore, it is necessary for us to research the development of novel antidepressants for effective treatment in practice. MicroRNA-204 (miR-204) is highly expressed in brain tissue, and plays a critical role in the synaptic plasticity of hippocampal neurons in rats. However, the underlying molecular mechanism of miR-204 remains unclear to date, this study aims to offer unique insights into depression and provide a theoretical basis for clinical physicians. METHODS A chronic social defeat stress (CSDS) was initially adopted for establishing a mice model of depression in this research and depression-like behaviors were evaluated by a series of behavioral experiments including the sucrose preference test (SPT), the tail suspension test (TST), the forced swim test (FST) and the social interaction test (SIT). Quantitative real-time reverse transcription PCR (qRT-PCR) was also conducted to test the expression levels of miR-204 and BDNF in the hippocampus of mice. Finally, gene interference of miR-204-5p was further adopted to test whether miR-204-5p played an effective role in the antidepressant effects of venlafaxine in mice. RESULTS Our data implicated that CSDS significantly increased the miR-204-5p but not miR-204-3p levels in the hippocampus of mice. The treatment of venlafaxine obviously relieved depression- like behaviors of CSDS-induced mice. The usage of venlafaxine abolished the increasing effects on the expression of miR-204-5p but up-regulated the BDNF expression level in CSDS-exposured mice. More importantly, we found that genetic overexpression of miR-204-5p decreased the reverse effects of venlafaxine on depressive-like behaviors and genetic knockdown of hippocampal miR-204-5p relieved the depressive-like behaviors and neurogenesis in CSDS-induced mice. CONCLUSION miR-204-5p played an effective role in the antidepressant effects of venlafaxine in CSDS-induced mice.
Collapse
Affiliation(s)
- Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong 226001, Jiangsu, China
| | - Xin-Yuan Wu
- Department of Gynaecology and Obstetrics, Yancheng Maternal and Child Health Care Hospital, Yancheng 224000 Jiangsu, China
| | - Xiang Jin
- Department of Pharmacy, The Second People's Hospital of Nantong, Nantong 226002, Jiangsu, China
| | - Xiao-Ming Sheng
- Department of Trauma Center, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Yan Fan
- Department of Pharmacy, Zhangjiagang Second People's Hospital, Zhangjiagang 215600, Jiangsu, China
| |
Collapse
|
3
|
Palamarchuk IS, Slavich GM, Vaillancourt T, Rajji TK. Stress-related cellular pathophysiology as a crosstalk risk factor for neurocognitive and psychiatric disorders. BMC Neurosci 2023; 24:65. [PMID: 38087196 PMCID: PMC10714507 DOI: 10.1186/s12868-023-00831-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/24/2023] [Indexed: 12/18/2023] Open
Abstract
In this narrative review, we examine biological processes linking psychological stress and cognition, with a focus on how psychological stress can activate multiple neurobiological mechanisms that drive cognitive decline and behavioral change. First, we describe the general neurobiology of the stress response to define neurocognitive stress reactivity. Second, we review aspects of epigenetic regulation, synaptic transmission, sex hormones, photoperiodic plasticity, and psychoneuroimmunological processes that can contribute to cognitive decline and neuropsychiatric conditions. Third, we explain mechanistic processes linking the stress response and neuropathology. Fourth, we discuss molecular nuances such as an interplay between kinases and proteins, as well as differential role of sex hormones, that can increase vulnerability to cognitive and emotional dysregulation following stress. Finally, we explicate several testable hypotheses for stress, neurocognitive, and neuropsychiatric research. Together, this work highlights how stress processes alter neurophysiology on multiple levels to increase individuals' risk for neurocognitive and psychiatric disorders, and points toward novel therapeutic targets for mitigating these effects. The resulting models can thus advance dementia and mental health research, and translational neuroscience, with an eye toward clinical application in cognitive and behavioral neurology, and psychiatry.
Collapse
Affiliation(s)
- Iryna S Palamarchuk
- Centre for Addiction and Mental Health, 1001 Queen Street West, Toronto, ON, M6J1H4, Canada.
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Sunnybrook Health Sciences Centre, Division of Neurology, Toronto, ON, Canada.
- Temerty Faculty of Medicine, Toronto Dementia Research Alliance, University of Toronto, Toronto, ON, Canada.
| | - George M Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tracy Vaillancourt
- Counselling Psychology, Faculty of Education, University of Ottawa, Ottawa, ON, Canada
- School of Psychology, Faculty of Social Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Tarek K Rajji
- Centre for Addiction and Mental Health, 1001 Queen Street West, Toronto, ON, M6J1H4, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, Toronto Dementia Research Alliance, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Lee J, Kim EJ, Park GS, Kim J, Kim TE, Lee YJ, Park J, Kang J, Koo JW, Choi TY. Lactobacillus reuteri ATG-F4 Alleviates Chronic Stress-induced Anhedonia by Modulating the Prefrontal Serotonergic System. Exp Neurobiol 2023; 32:313-327. [PMID: 37927130 PMCID: PMC10628864 DOI: 10.5607/en23028] [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: 09/04/2023] [Revised: 10/02/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023] Open
Abstract
Mental health is influenced by the gut-brain axis; for example, gut dysbiosis has been observed in patients with major depressive disorder (MDD). Gut microbial changes by fecal microbiota transplantation or probiotics treatment reportedly modulates depressive symptoms. However, it remains unclear how gut dysbiosis contributes to mental dysfunction, and how correction of the gut microbiota alleviates neuropsychiatric disorders. Our previous study showed that chronic consumption of Lactobacillus reuteri ATG-F4 (F4) induced neurometabolic alterations in healthy mice. Here, we investigated whether F4 exerted therapeutic effects on depressive-like behavior by influencing the central nervous system. Using chronic unpredictable stress (CUS) to induce anhedonia, a key symptom of MDD, we found that chronic F4 consumption alleviated CUS-induced anhedonic behaviors, accompanied by biochemical changes in the gut, serum, and brain. Serum and brain metabolite concentrations involved in tryptophan metabolism were regulated by CUS and F4. F4 consumption reduced the elevated levels of serotonin (5-HT) in the brain observed in the CUS group. Additionally, the increased expression of Htr1a, a subtype of the 5-HT receptor, in the medial prefrontal cortex (mPFC) of stressed mice was restored to levels observed in stress-naïve mice following F4 supplementation. We further demonstrated the role of Htr1a using AAV-shRNA to downregulate Htr1a in the mPFC of CUS mice, effectively reversing CUS-induced anhedonic behavior. Together, our findings suggest F4 as a potential therapeutic approach for relieving some depressive symptoms and highlight the involvement of the tryptophan metabolism in mitigating CUS-induced depressive-like behaviors through the action of this bacterium.
Collapse
Affiliation(s)
- Jiyun Lee
- Emotion, Cognition, and Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Eum-Ji Kim
- Emotion, Cognition, and Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| | | | - Jeongseop Kim
- Emotion, Cognition, and Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Tae-Eun Kim
- Emotion, Cognition, and Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Yoo Jin Lee
- Emotion, Cognition, and Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| | - Juyi Park
- AtoGen Co., Ltd., Daejeon 34015, Korea
| | | | - Ja Wook Koo
- Emotion, Cognition, and Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Tae-Yong Choi
- Emotion, Cognition, and Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| |
Collapse
|
5
|
Zhou R, Wang G, Li Q, Meng F, Liu C, Gan R, Ju D, Liao M, Xu J, Sang D, Gao X, Zhou S, Wu K, Sun Q, Guo Y, Wu C, Chen Z, Chen L, Shi B, Wang H, Wang X, Li H, Cai T, Li B, Wang F, Funato H, Yanagisawa M, Zhang EE, Liu Q. A signalling pathway for transcriptional regulation of sleep amount in mice. Nature 2022; 612:519-527. [PMID: 36477534 DOI: 10.1038/s41586-022-05510-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/01/2022] [Indexed: 12/12/2022]
Abstract
In mice and humans, sleep quantity is governed by genetic factors and exhibits age-dependent variation1-3. However, the core molecular pathways and effector mechanisms that regulate sleep duration in mammals remain unclear. Here, we characterize a major signalling pathway for the transcriptional regulation of sleep in mice using adeno-associated virus-mediated somatic genetics analysis4. Chimeric knockout of LKB1 kinase-an activator of AMPK-related protein kinase SIK35-7-in adult mouse brain markedly reduces the amount and delta power-a measure of sleep depth-of non-rapid eye movement sleep (NREMS). Downstream of the LKB1-SIK3 pathway, gain or loss-of-function of the histone deacetylases HDAC4 and HDAC5 in adult brain neurons causes bidirectional changes of NREMS amount and delta power. Moreover, phosphorylation of HDAC4 and HDAC5 is associated with increased sleep need, and HDAC4 specifically regulates NREMS amount in posterior hypothalamus. Genetic and transcriptomic studies reveal that HDAC4 cooperates with CREB in both transcriptional and sleep regulation. These findings introduce the concept of signalling pathways targeting transcription modulators to regulate daily sleep amount and demonstrate the power of somatic genetics in mouse sleep research.
Collapse
Affiliation(s)
- Rui Zhou
- College of Biological Sciences, China Agriculture University, Beijing, China
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Guodong Wang
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qi Li
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), Tsinghua University, Beijing, China
| | - Fanxi Meng
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Can Liu
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
- Peking University-Tsinghua University-NIBS Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, China
| | - Rui Gan
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Dapeng Ju
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Meimei Liao
- College of Biological Sciences, China Agriculture University, Beijing, China
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Junjie Xu
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
- College of Life Sciences, Beijing Normal University, Beijing, China
| | - Di Sang
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xue Gao
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Shuang Zhou
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
- College of Life Sciences, Beijing Normal University, Beijing, China
| | - Kejia Wu
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Quanzhi Sun
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Ying Guo
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Chongyang Wu
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Zhiyu Chen
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Lin Chen
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Bihan Shi
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Haiyan Wang
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Xia Wang
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Huaiye Li
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Tao Cai
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), Tsinghua University, Beijing, China
| | - Bin Li
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
| | - Fengchao Wang
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), Tsinghua University, Beijing, China
| | - Hiromasa Funato
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Eric Erquan Zhang
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), Tsinghua University, Beijing, China
| | - Qinghua Liu
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), Tsinghua University, Beijing, China.
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
6
|
Long Noncoding RNA LINC00473 Ameliorates Depression-Like Behaviors in Female Mice by Acting as a Molecular Sponge to Regulate miR-497-5p/BDNF Axis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:4244425. [PMID: 36072768 PMCID: PMC9441382 DOI: 10.1155/2022/4244425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/28/2022] [Accepted: 08/08/2022] [Indexed: 11/18/2022]
Abstract
Background. Depression was a common life-threatening psychiatric disorder and occurs more frequently in women than in men. Long noncoding RNAs (lncRNAs), such as LINC00473, had been reported to be involved in the progression of depression. Methods. Chronic unpredictable moderate stress in mice (CUMS) was applied to construct a depression model. Subsequently, RT-qPCR was applied to check the level of LINC00473 and microRNA-497-5p (miR-497-5p) in the hippocampal region of the mice induced by CUMS. CUMS mice were injected with lentiviral vectors of LINC00473 (LV-LINC00473), miR-497-5p inhibitor, short hairpin- (sh-) brain-derived neurotrophic factor (sh-BDNF), or miR-497-5p mimic to evaluate depressive behaviors, including sucrose preference test, forced swim test, elevated plus maze, and tail suspension test. Moreover, the production of hypothalamic neurotransmitters was assessed with the usage of ELISA kits. Dual-luciferase reporter assay, RNA pull-down, and RIP analysis were performed to measure the relationship between miR-497-5p and LINC00473 or BDNF. Further, western blot was employed to determine the protein level of BDNF. Results. We discovered that LINC00473 level was downregulated in the female mice with depression, but not in male mice. Besides, the depressive behaviors induced by CUMS in mice, including the decrease of sucrose preference and time in open arm, as well as the increase of immobility time and swimming resting time were all ameliorated by LINC00473 overexpression. Moreover, the concentration of neurotransmitters was decreased in CUMS-induced mouse hypothalamus, which was blocked by LV-LINC00473 lentiviral vector administration. Mechanistically, LINC00473 directly targeted miR-497-5p. Absence of miR-497-5p revealed the antidepression effects on CUMS-induced mice, and miR-497-5p upregulation could counter the antidepressive impacts of LINC00473 upregulation on CUMS-induced mice. Furthermore, LINC00473 could target miR-497-5p to modulate BDNF level. Knockdown of BDNF could abrogate the improving influences of miR-497-5p suppression on CUMS-induced depression. Conclusions. LINC00473 ameliorated CUMS-caused depression by encouraging BDNF expression via binding to miR-497-5p, which might provide a potential therapeutic target for depression in females.
Collapse
|
7
|
Shi LS, Ji CH, Tang WQ, Liu Y, Zhang W, Guan W. Hippocampal miR-124 Participates in the Pathogenesis of Depression via Regulating the Expression of BDNF in a Chronic Social Defeat Stress Model of Depression. Curr Neurovasc Res 2022; 19:210-218. [PMID: 35838216 DOI: 10.2174/1567202619666220713105306] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 01/27/2023]
Abstract
OBJECTIVE As one of the most prevalent psychiatric disorders, the exact pathogenesis of depression remains elusive. Therefore, there is an urgent need to identify novel antidepressants for effective treatment. MicroRNA-124 (miR-124), the most abundant miRNA in brain tissue, plays a key effect on adult neurogenesis and neuronal differentiation. However, the mechanism of miR-124 in depression has not been clarified so far. The aim of this study is to provide broad insight into the mechanisms underlying depression. METHODS In the study, we used the forced swim test (FST), the tail suspension test (TST), and a Chronic Social Defeat Stress (CSDS) mice model of depression. Quantitative real-time reverse transcription PCR (qRT-PCR), western blotting, immunofluorescence and virus-mediated gene transfer were used together. The level of plasma corticosterone in mice was analyzed by Enzyme Linked Immunosorbent Assay (ELISA). RESULTS It was found that CSDS robustly increased the level of miR-124 in the hippocampus. Genetic knockdown of hippocampal miR-124 produced significant antidepressant-like effects in the CSDS model of depression. Furthermore, AAV-siR-124-EGFP treatment increased the level of plasma corticosterone in CSDS-induced mice. Moreover, it was found that the antidepressant-like effects induced by miR-124 inhibition required the hippocampal BDNF-TrkB system. CONCLUSION Hippocampal miR-124 participated in the pathogenesis of depression by regulating BDNF biosynthesis and was a feasible antidepressant target.
Collapse
Affiliation(s)
- Lin-Sheng Shi
- Department of Cardiology, Affiliated Hospital 2 of Nantong University, Nantong 226001, Jiangsu, China
| | - Chun-Hui Ji
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong 226001, Jiangsu, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong 226001, Jiangsu, China
| | - Wen-Qian Tang
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong 226001, Jiangsu, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong 226001, Jiangsu, China
| | - Yue Liu
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong 226001, Jiangsu, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong 226001, Jiangsu, China
| | - Wei Zhang
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong 226001, Jiangsu, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong 226001, Jiangsu, China
| | - Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong 226001, Jiangsu, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong 226001, Jiangsu, China
| |
Collapse
|
8
|
Su Y, Wang W, Meng X. Revealing the Roles of MOAP1 in Diseases: A Review. Cells 2022; 11:cells11050889. [PMID: 35269511 PMCID: PMC8909730 DOI: 10.3390/cells11050889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
Modulator of apoptosis protein1 (MOAP1), also known as MAP1 and PNMA4, belongs to the PNMA gene family consisting of at least 15 genes located on different chromosomes. MOAP1 interacts with the BAX protein, one of the most important apoptosis regulators. Due to its critical role in a few of disease-associated pathways, MOAP1 is associated with many diseases such as cancers and neurological diseases. In this study, we introduced MOAP1 and its biological functions and reviewed the associations between MOAP1 and a few diseases including cancers, neurological diseases, and other diseases such as inflammation and heart diseases. We also explained possible biological mechanisms underlying the associations between MOAP1 and these diseases, and discussed a few future directions regarding MOAP1, especially its potential roles in neurodegenerative disorders. In summary, MOAP1 plays a critical role in the development and progression of cancers and neurological diseases by regulating a few genes related to cellular apoptosis such as BAX and RASSF1A and interacting with disease-associated miRNAs, including miR-25 and miR1228.
Collapse
|
9
|
Yang W, Liu M, Zhang Q, Zhang J, Chen J, Chen Q, Suo L. Knockdown of miR-124 Reduces Depression-like Behavior by Targeting CREB1 and BDNF. Curr Neurovasc Res 2020; 17:196-203. [PMID: 32189593 PMCID: PMC7536798 DOI: 10.2174/1567202617666200319141755] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 01/22/2020] [Accepted: 01/25/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE As a brain-specific microRNA, the mechanism of miR-124 in depression has not been clarified so far. The present study aimed to explore the role of miR-124 in depression and its potential targets. METHODS The depression model was first replicated by the chronic unpredictable mild stress (CUMS) method. miR-124 antagomir was injected into the hippocampus of CUMS rats. Sucrose preference test (SPT), open-field test (OFT), elevated-plus maze (EPM), and forced swimming test (FST) were used to analyze the depression-like behavior. The content of norepinephrine (NE), dopamine (DA) and 5-hydroxytryptamine (5-HT) in the hypothalamus was analyzed by ELISA. qRT-PCR and western blot assay were used for functional analysis. RESULTS miR-124 expression was up-regulated in the hippocampus of CUMS -induced depression model rats, while CREB1 and BDNF were down-regulated. Administration of miR-124 antagomir in the hippocampus inhibited miR-124 expression in the hippocampus of CUMS rats. Additionally, SPT, OFT, EPM, and FST also showed that miR-124 antagomir can reduce the depression-like behavior of CUMS rats. Furthermore, miR-124 antagomir injection increased the levels of NE, DA and 5-HT in the hypothalamus of CUMS rats. Moreover, miR-124 antagomir injection increased the expression of cyclic AMP-responsive element-binding protein1 (CREB1) and brain-derived neurotrophic factor (BDNF) in the hippocampus. Using the dual-luciferase reporter assay, it was confirmed that miR-124 directly targets 3'UTR of CREB1 and BDNF genes. CONCLUSION Knockdown of miR-124 can improve depression-like behavior in CUMS-induced depressive rats, which may be related at least in part to the up-regulation of CREB1 and BDNF expression in the hippocampus.
Collapse
Affiliation(s)
- Wei Yang
- Department of Endocrinology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Min Liu
- Department of Endocrinology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Qianwei Zhang
- Department of Endocrinology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Jiahua Zhang
- Department of Endocrinology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Jun Chen
- Department of Endocrinology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Qiaoyun Chen
- Department of Endocrinology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Lixia Suo
- Department of Endocrinology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|