1
|
Ramirez-Gomez J, Dalal S, Devara D, Sharma B, Rodarte D, Kumar S. MicroRNA-based recent research developments in Alzheimer's disease. J Alzheimers Dis 2025; 104:14-31. [PMID: 39894921 DOI: 10.1177/13872877241313397] [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: 02/04/2025]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder that is characterized by memory and physical impairment in aged individuals. microRNAs (miRNAs) are small, single-stranded noncoding RNAs that induce translational repression by binding to the 3' UTR of a target mRNA. miRNAs play a crucial role in neurological activity by mediating cellular proliferation, synaptic plasticity, apoptosis and more. Ongoing research in patents and clinical trials have called attention to promising miRNAs as biomarkers and therapeutics in AD. Recent research has shown that miRNAs are aberrantly expressed in AD brain, blood, cerebrospinal fluid and serum. Attenuated miRNA expressions have diagnostic potential in AD by interacting with amyloid-β synthesis, phosphorylated tau, and neurofibrillary tangles. In this study, miRNA-29a, miRNA-125b, miRNA-34a, miRNA-146a, and miRNA-155 have shown promise as potential biomarker candidates for AD. Improving cognitive symptoms can be traced to restoring the endogenous miRNA activity by synthesizing miRNA mimics and miRNA antisense oligonucleotides. miRNA-483-5p, miRNA-188-5p, miRNA-219, miRNA135a/5p, miRNA-23/23b-3p, miRNA-124, and miRNA-455-3p are growing therapeutics for AD. However, miRNA-based therapeutics struggle outside of preclinical testing. miRNA-107, miRNA-206, miRNA-30/7, and miRNA-142-3p face bottlenecks in clinical trials due to a lack of experimental design, transparency and volunteer size. Patenting recent miRNA-based developments demonstrates the commitment in identifying a new biomarker and/or therapeutic for AD.
Collapse
Affiliation(s)
- Jaime Ramirez-Gomez
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Sarthak Dalal
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Davin Devara
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Bhupender Sharma
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Daniela Rodarte
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Subodh Kumar
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
- L. Frederick Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX, USA
| |
Collapse
|
2
|
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 2025; 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] [MESH Headings] [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
|
3
|
Parkins EV, Gross C. Small Differences and Big Changes: The Many Variables of MicroRNA Expression and Function in the Brain. J Neurosci 2024; 44:e0365242024. [PMID: 39111834 PMCID: PMC11308354 DOI: 10.1523/jneurosci.0365-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 08/10/2024] Open
Abstract
MicroRNAs are emerging as crucial regulators within the complex, dynamic environment of the synapse, and they offer a promising new avenue for the treatment of neurological disease. These small noncoding RNAs modify gene expression in several ways, including posttranscriptional modulation via binding to complementary and semicomplementary sites on target mRNAs. This rapid, finely tuned regulation of gene expression is essential to meet the dynamic demands of the synapse. Here, we provide a detailed review of the multifaceted world of synaptic microRNA regulation. We discuss the many mechanisms by which microRNAs regulate gene expression at the synapse, particularly in the context of neuronal plasticity. We also describe the various factors, such as age, sex, and neurological disease, that can influence microRNA expression and activity in neurons. In summary, microRNAs play a crucial role in the intricate and quickly changing functional requirements of the synapse, and context is essential in the study of microRNAs and their potential therapeutic applications.
Collapse
Affiliation(s)
- Emma V Parkins
- University of Cincinnati Neuroscience Graduate Program, Cincinnati, Ohio 45229
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Christina Gross
- University of Cincinnati Neuroscience Graduate Program, Cincinnati, Ohio 45229
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229
| |
Collapse
|
4
|
Dalal S, Ramirez-Gomez J, Sharma B, Devara D, Kumar S. MicroRNAs and synapse turnover in Alzheimer's disease. Ageing Res Rev 2024; 99:102377. [PMID: 38871301 DOI: 10.1016/j.arr.2024.102377] [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: 04/23/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of amyloid-beta plaques and neurofibrillary tangles in the brain, leading to synaptic dysfunction and cognitive decline. Healthy synapses are the crucial for normal brain function, memory restoration and other neurophysiological function. Synapse loss and synaptic dysfunction are two primary events that occur during AD initiation. Synapse lifecycle and/or synapse turnover is divided into five key stages and several sub-stages such as synapse formation, synapse assembly, synapse maturation, synapse transmission and synapse termination. In normal state, the synapse turnover is regulated by various biological and molecular factors for a healthy neurotransmission. In AD, the different stages of synapse turnover are affected by AD-related toxic proteins. MicroRNAs (miRNAs) have emerged as critical regulators of gene expression and have been implicated in various neurological diseases, including AD. Deregulation of miRNAs modulate the synaptic proteins and affect the synapse turnover at different stages. In this review, we discussed the key milestones of synapse turnover and how they are affected in AD. Further, we discussed the involvement of miRNAs in synaptic turnover, focusing specifically on their role in AD pathogenesis. We also emphasized the regulatory mechanisms by which miRNAs modulate the synaptic turnover stages in AD. Current studies will help to understand the synaptic life-cycle and role of miRNAs in each stage that is deregulated in AD, further allowing for a better understanding of the pathogenesis of devastating disease.
Collapse
Affiliation(s)
- Sarthak Dalal
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Jaime Ramirez-Gomez
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Bhupender Sharma
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Davin Devara
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Subodh Kumar
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA; L. Frederick Francis Graduate School of Biomedicael Sciences, Texas Tech University Health Sciences Center, El Paso, TX, USA.
| |
Collapse
|
5
|
Park I, Kim HJ, Shin J, Jung YJ, Lee D, Lim J, Park JM, Park JW, Kim J. AFM Imaging Reveals MicroRNA-132 to be a Positive Regulator of Synaptic Functions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306630. [PMID: 38493494 PMCID: PMC11077659 DOI: 10.1002/advs.202306630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/17/2024] [Indexed: 03/19/2024]
Abstract
The modification of synaptic and neural connections in adults, including the formation and removal of synapses, depends on activity-dependent synaptic and structural plasticity. MicroRNAs (miRNAs) play crucial roles in regulating these changes by targeting specific genes and regulating their expression. The fact that somatic and dendritic activity in neurons often occurs asynchronously highlights the need for spatial and dynamic regulation of protein synthesis in specific milieu and cellular loci. MicroRNAs, which can show distinct patterns of enrichment, help to establish the localized distribution of plasticity-related proteins. The recent study using atomic force microscopy (AFM)-based nanoscale imaging reveals that the abundance of miRNA(miR)-134 is inversely correlated with the functional activity of dendritic spine structures. However, the miRNAs that are selectively upregulated in potentiated synapses, and which can thereby support prospective changes in synaptic efficacy, remain largely unknown. Using AFM force imaging, significant increases in miR-132 in the dendritic regions abutting functionally-active spines is discovered. This study provides evidence for miR-132 as a novel positive miRNA regulator residing in dendritic shafts, and also suggests that activity-dependent miRNAs localized in distinct sub-compartments of neurons play bi-directional roles in controlling synaptic transmission and synaptic plasticity.
Collapse
Affiliation(s)
- Ikbum Park
- Technical Support Center for Chemical IndustryKorea Research Institute of Chemical Technology (KRICT)Ulsan44412Republic of Korea
| | - Hyun Jin Kim
- Department of Life SciencesPohang University of Science and Technology (POSTECH)Pohang37673Republic of Korea
| | - Juyoung Shin
- Department of Life SciencesPohang University of Science and Technology (POSTECH)Pohang37673Republic of Korea
| | - Yu Jin Jung
- Center for Specialty ChemicalsKorea Research Institute of Chemical Technology (KRICT)Ulsan44412Republic of Korea
| | - Donggyu Lee
- Division of Electronics and Information SystemDaegu Gyeongbuk Institute of Science and Technology (DGIST)Daegu42988Republic of Korea
| | - Ji‐seon Lim
- Department of ChemistryPohang University of Science and Technology (POSTECH)Pohang37673Republic of Korea
| | - Jong Mok Park
- Technical Support Center for Chemical IndustryKorea Research Institute of Chemical Technology (KRICT)Ulsan44412Republic of Korea
| | - Joon Won Park
- Department of ChemistryPohang University of Science and Technology (POSTECH)Pohang37673Republic of Korea
| | - Joung‐Hun Kim
- Department of Life SciencesPohang University of Science and Technology (POSTECH)Pohang37673Republic of Korea
- Institute of Convergence ScienceYonsei UniversitySeoul03722Republic of Korea
| |
Collapse
|
6
|
Kaurani L. Clinical Insights into MicroRNAs in Depression: Bridging Molecular Discoveries and Therapeutic Potential. Int J Mol Sci 2024; 25:2866. [PMID: 38474112 PMCID: PMC10931847 DOI: 10.3390/ijms25052866] [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: 01/31/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Depression is a major contributor to the overall global burden of disease. The discovery of biomarkers for diagnosis or prediction of treatment responses and as therapeutic agents is a current priority. Previous studies have demonstrated the importance of short RNA molecules in the etiology of depression. The most extensively researched of these are microRNAs, a major component of cellular gene regulation and function. MicroRNAs function in a temporal and tissue-specific manner to regulate and modify the post-transcriptional expression of target mRNAs. They can also be shuttled as cargo of extracellular vesicles between the brain and the blood, thus informing about relevant mechanisms in the CNS through the periphery. In fact, studies have already shown that microRNAs identified peripherally are dysregulated in the pathological phenotypes seen in depression. Our article aims to review the existing evidence on microRNA dysregulation in depression and to summarize and evaluate the growing body of evidence for the use of microRNAs as a target for diagnostics and RNA-based therapies.
Collapse
Affiliation(s)
- Lalit Kaurani
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany
| |
Collapse
|
7
|
Abdel-Reheim MA, Nomier Y, Zaki MB, Abulsoud AI, Mohammed OA, Rashad AA, Oraby MA, Elballal MS, Tabaa MME, Elazazy O, Abd-Elmawla MA, El-Dakroury WA, Abdel Mageed SS, Abdelmaksoud NM, Elrebehy MA, Helal GK, Doghish AS. Unveiling the regulatory role of miRNAs in stroke pathophysiology and diagnosis. Pathol Res Pract 2024; 253:155085. [PMID: 38183822 DOI: 10.1016/j.prp.2023.155085] [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: 12/14/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/08/2024]
Abstract
Stroke, a major global cause of mortality, leads to a range of problems for those who survive. Besides its brutal events, stroke also tends to have a characteristic of recurrence, making it a complex disease involving intricate regulatory networks. One of the major cellular regulators is the non-coding RNAs (ncRNA), specifically microRNAs (miRNAs), thus the possible functions of miRNAs in the pathogenesis of stroke are discussed as well as the possibility of using miRNA-based therapeutic approaches. Firstly, the molecular mechanisms by which miRNAs regulate vital physiological processes, including synaptic plasticity, oxidative stress, apoptosis, and the integrity of the blood-brain barrier (BBB) are reviewed. The miRNA indirectly impacts stroke outcomes by regulating BBB function and angiogenesis through the targeting of transcription factors and angiogenic factors. In addition, the tendency for some miRNAs to be upregulated in response to hypoxia, which is a prevalent phenomenon in stroke and various neurological disorders, highlights the possibility that it controls hypoxia-inducible factor (HIF) signaling and angiogenesis, thereby influencing the integrity of the BBB as examples of the discussed mechanisms. Furthermore, this review explores the potential therapeutic targets that miRNAs may offer for stroke recovery and highlights their promising capacity to alleviate post-stroke complications. This review provides researchers and clinicians with valuable resources since it attempts to decipher the complex network of miRNA-mediated mechanisms in stroke. Additionally, the review addresses the interplay between miRNAs and stroke risk factors as well as clinical applications of miRNAs as diagnostic and prognostic markers.
Collapse
Affiliation(s)
- Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Yousra Nomier
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and health sciences, Sultan Qaboos University, Muscat, Oman
| | - Mohamed Bakr Zaki
- Biochemistry, Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menoufia 32897, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Ahmed A Rashad
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mamdouh A Oraby
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Manar Mohammed El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute (ESRI), University of Sadat City, Sadat City 32897, Menoufia, Egypt
| | - Ola Elazazy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mai A Abd-Elmawla
- Biochemistry, Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | | | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Gouda Kamel Helal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 11231, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| |
Collapse
|
8
|
Feng DD, Chen JH, Chen YF, Cao Q, Li BJ, Chen XQ, Jin R, Zhou GP. MALAT1 binds to miR-188-3p to regulate ALOX5 activity in the lung inflammatory response of neonatal bronchopulmonary dysplasia. Mol Immunol 2023; 160:67-79. [PMID: 37385102 DOI: 10.1016/j.molimm.2023.06.008] [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: 01/28/2023] [Revised: 04/23/2023] [Accepted: 06/19/2023] [Indexed: 07/01/2023]
Abstract
Bronchopulmonary dysplasia (BPD) causes high morbidity and mortality in infants, but no effective preventive or therapeutic agents have been developed to combat BPD. In this study, we assessed the expression of MALAT1 and ALOX5 in peripheral blood mononuclear cells from BPD neonates, hyperoxia-induced rat models and lung epithelial cell lines. Interestingly, we found upregulated expression of MALAT1 and ALOX5 in the experimental groups, along with upregulated expression of proinflammatory cytokines. According to bioinformatics prediction, MALAT1 and ALOX5 simultaneously bind to miR-188-3p, which was downregulated in the experimental groups above. Silencing MALAT1 or ALOX5 and overexpressing miR-188-3p inhibited apoptosis and promoted the proliferation of hyperoxia-treated A549 cells. Suppressing MALAT1 or overexpressing miR-188-3p increased the expression levels of miR-188-3p but decreased the expression levels of ALOX5. Moreover, RNA immunoprecipitation (RIP) and luciferase assays showed that MALAT1 directly targeted miR-188-3p to regulate ALOX5 expression in BPD neonates. Collectively, our study demonstrates that MALAT1 regulates ALOX5 expression by binding to miR-188-3p, providing novel insights into potential therapeutics for BPD treatment.
Collapse
Affiliation(s)
- Dan-Dan Feng
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Jia-He Chen
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Yu-Fei Chen
- Department of Pediatrics, Yancheng Maternal and Child Health Care Hospital, Yancheng 224000, China
| | - Qian Cao
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Bing-Jie Li
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Xiao-Qing Chen
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Rui Jin
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Guo-Ping Zhou
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
9
|
Koropouli E, Wang Q, Mejías R, Hand R, Wang T, Ginty DD, Kolodkin AL. Palmitoylation regulates neuropilin-2 localization and function in cortical neurons and conveys specificity to semaphorin signaling via palmitoyl acyltransferases. eLife 2023; 12:e83217. [PMID: 37010951 PMCID: PMC10069869 DOI: 10.7554/elife.83217] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 02/22/2023] [Indexed: 04/04/2023] Open
Abstract
Secreted semaphorin 3F (Sema3F) and semaphorin 3A (Sema3A) exhibit remarkably distinct effects on deep layer excitatory cortical pyramidal neurons; Sema3F mediates dendritic spine pruning, whereas Sema3A promotes the elaboration of basal dendrites. Sema3F and Sema3A signal through distinct holoreceptors that include neuropilin-2 (Nrp2)/plexinA3 (PlexA3) and neuropilin-1 (Nrp1)/PlexA4, respectively. We find that Nrp2 and Nrp1 are S-palmitoylated in cortical neurons and that palmitoylation of select Nrp2 cysteines is required for its proper subcellular localization, cell surface clustering, and also for Sema3F/Nrp2-dependent dendritic spine pruning in cortical neurons, both in vitro and in vivo. Moreover, we show that the palmitoyl acyltransferase ZDHHC15 is required for Nrp2 palmitoylation and Sema3F/Nrp2-dependent dendritic spine pruning, but it is dispensable for Nrp1 palmitoylation and Sema3A/Nrp1-dependent basal dendritic elaboration. Therefore, palmitoyl acyltransferase-substrate specificity is essential for establishing compartmentalized neuronal structure and functional responses to extrinsic guidance cues.
Collapse
Affiliation(s)
- Eleftheria Koropouli
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Qiang Wang
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Rebeca Mejías
- Department of Physiology,University of SevilleSevilleSpain
| | - Randal Hand
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Tao Wang
- McKusick-Nathans Institute of Genetic Medicine and Department of Pediatrics, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Alex L Kolodkin
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
10
|
Roy B, Dwivedi Y. An insight into the sprawling microverse of microRNAs in depression pathophysiology and treatment response. Neurosci Biobehav Rev 2023; 146:105040. [PMID: 36639069 PMCID: PMC9974865 DOI: 10.1016/j.neubiorev.2023.105040] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/12/2023]
Abstract
Stress-related neuropathologies are pivotal in developing major depressive disorder (MDD) and are often governed by gene-regulatory changes. Being a stress-responsive gene-regulatory factor, microRNAs (miRNAs) have tremendous biomolecular potential to define an altered gene-regulatory landscape in the MDD brain. MiRNAs' regulatory roles in the MDD brain are closely aligned with changes in plasticity, neurogenesis, and stress-axis functions. MiRNAs act at the epigenetic interface between stress-induced environmental stimuli and cellular pathologies by triggering large-scale gene expression changes in a highly coordinated fashion. The parallel changes in peripheral circulation may provide an excellent opportunity for miRNA to devise more effective treatment strategies and help explore their potential as biomarkers in treatment response. This review discusses the role of miRNAs as epigenetic modifiers in the etiopathogenesis of MDD. Concurrently, key research is highlighted to show the progress in using miRNAs as predictive biomarkers for treatment response.
Collapse
Affiliation(s)
- Bhaskar Roy
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
11
|
Long-term potentiation and depression regulatory microRNAs were highlighted in Bisphenol A induced learning and memory impairment by microRNA sequencing and bioinformatics analysis. PLoS One 2023; 18:e0279029. [PMID: 36656826 PMCID: PMC9851566 DOI: 10.1371/journal.pone.0279029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 11/28/2022] [Indexed: 01/20/2023] Open
Abstract
The mechanisms of Bisphenol A (BPA) induced learning and memory impairment have still not been fully elucidated. MicroRNAs (miRNAs) are endogenous non-coding small RNA molecules involved in the process of toxicant-induced neurotoxicity. To investigate the role of miRNAs in BPA-induced learning and memory impairment, we analyzed the impacts of BPA on miRNA expression profile by high-throughput sequencing in mice hippocampus. Results showed that mice treated with BPA displayed impairments of spatial learning and memory and changes in the expression of miRNAs in the hippocampus. Seventeen miRNAs were significantly differentially expressed after BPA exposure, of these, 13 and 4 miRNAs were up- and downregulated, respectively. Bioinformatic analysis of Gene Ontology (GO) and pathway suggests that BPA exposure significantly triggered transcriptional changes of miRNAs associated with learning and memory; the top five affected pathways involved in impairment of learning and memory are: 1) Long-term depression (LTD); 2) Thyroid hormone synthesis; 3) GnRH signaling pathway; 4) Long-term potentiation (LTP); 5) Serotonergic synapse. Eight BPA-responsive differentially expressed miRNAs regulating LTP and LTD were further screened to validate the miRNA sequencing data using Real-Time PCR. The deregulation expression levels of proteins of five target genes (CaMKII, MEK1/2, IP3R, AMPAR1 and PLCβ4) were investigated via western blot, for further verifying the results of gene target analysis. Our results showed that LTP and LTD related miRNAs and their targets could contribute to BPA-induced impairment of learning and memory. This study provides valuable information for novel miRNA biomarkers to detect changes in impairment of learning and memory induced by BPA exposure.
Collapse
|
12
|
Mohammadi AH, Seyedmoalemi S, Moghanlou M, Akhlagh SA, Talaei Zavareh SA, Hamblin MR, Jafari A, Mirzaei H. MicroRNAs and Synaptic Plasticity: From Their Molecular Roles to Response to Therapy. Mol Neurobiol 2022; 59:5084-5102. [PMID: 35666404 DOI: 10.1007/s12035-022-02907-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 05/25/2022] [Indexed: 12/24/2022]
Abstract
Synaptic plasticity is the ability of synapses to weaken or strengthen over time, in response to changes in the activity of the neurons. It is orchestrated by a variety of genes, proteins, and external and internal factors, especially epigenetic factors. MicroRNAs (miRNAs) are well-acknowledged epigenetic modulators that regulate the translation and degradation of target genes in the nervous system. Increasing evidence has suggested that a number of miRNAs play important roles in modulating various aspects of synaptic plasticity. The deregulation of miRNAs could be associated with pathological alterations in synaptic plasticity, which could lead to different CNS-related diseases. Herein, we provide an update on the role of miRNAs in governing synaptic plasticity. In addition, we also summarize recent researches on the role of miRNAs in drug addiction, and their targets and mechanism of action. Understanding of the way in which miRNAs contribute to synaptic plasticity provides rational clues in establishing the novel biomarkers and new therapeutic strategies for the diagnosis and treatment of plasticity-related diseases and drug addiction.
Collapse
Affiliation(s)
- Amir Hossein Mohammadi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyedvahid Seyedmoalemi
- Behavioral Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahsa Moghanlou
- Department of Psychiatry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Ameneh Jafari
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
13
|
Gao YN, Zhang YQ, Wang H, Deng YL, Li NM. A New Player in Depression: MiRNAs as Modulators of Altered Synaptic Plasticity. Int J Mol Sci 2022; 23:ijms23094555. [PMID: 35562946 PMCID: PMC9101307 DOI: 10.3390/ijms23094555] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 01/04/2023] Open
Abstract
Depression is a psychiatric disorder that presents with a persistent depressed mood as the main clinical feature and is accompanied by cognitive impairment. Changes in neuroplasticity and neurogenesis greatly affect depression. Without genetic changes, epigenetic mechanisms have been shown to function by regulating gene expression during the body’s adaptation to stress. Studies in recent years have shown that as important regulatory factors in epigenetic mechanisms, microRNAs (miRNAs) play important roles in the development and progression of depression through the regulation of protein expression. Herein, we review the mechanisms of miRNA-mediated neuroplasticity in depression and discus synaptic structural plasticity, synaptic functional plasticity, and neurogenesis. Furthermore, we found that miRNAs regulate neuroplasticity through several signalling pathways to affect cognitive functions. However, these pathways do not work independently. Therefore, we try to identify synergistic correlations between miRNAs and multiple signalling pathways to broaden the potential pathogenesis of depression. In addition, in the future, dual-function miRNAs (protection/injury) are promising candidate biomarkers for the diagnosis of depression, and their regulated genes can potentially be used as target genes for the treatment of depression.
Collapse
Affiliation(s)
- Ya-Nan Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China; (Y.-N.G.); (H.W.)
| | - Yong-Qian Zhang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (Y.-Q.Z.); (Y.-L.D.)
| | - Hao Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China; (Y.-N.G.); (H.W.)
| | - Yu-Lin Deng
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (Y.-Q.Z.); (Y.-L.D.)
| | - Nuo-Min Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China; (Y.-N.G.); (H.W.)
- Correspondence:
| |
Collapse
|
14
|
Non-coding RNA in alcohol use disorder by affecting synaptic plasticity. Exp Brain Res 2022; 240:365-379. [PMID: 35028694 DOI: 10.1007/s00221-022-06305-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 01/04/2022] [Indexed: 12/19/2022]
Abstract
Alcohol use disorder (AUD) is one of the most serious public health problems worldwide. AUD is a complex disorder, and there is ample evidence that genetic predisposition is critical to its development. Recent studies have shown that genetic predisposition leads to the onset of AUD, and alcohol metabolism can affect epigenetic inheritance, which in turn affects synaptic plasticity, alters brain function, and leads to more severe addictive behaviors. Non-coding RNAs (ncRNAs), especially microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), play an important role in alcohol addiction. This paper reviews the regulatory role of ncRNAs. ncRNAs are involved in enzyme and neurotransmitter reaction systems during alcohol use disorder. Alcohol consumption regulates the expression of ncRNAs that mediate epigenetic modification and synaptic plasticity, which play an important role in the development of chronic AUD. ncRNAs may be used not only as predictors of therapeutic responses but also as therapeutic targets of AUD. Chronic alcoholism is more likely to lead to neuroimmune disorders, including permanent brain dysfunction. AUD induced by long-term alcoholism greatly alters the expression of genes in the human genome, especially the expression of ncRNAs. Alcohol can cause a series of pathological changes by interfering with gene expression, such as through disordered miRNA-mRNA expression networks, epigenetic modifications, disordered metabolism, and even synaptic remodeling. ncRNAs are involved in the transition from moderate drinking to alcohol dependence.
Collapse
|
15
|
MicroRNA Dysregulation in the Hippocampus of Rats with Noise-Induced Hearing Loss. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1377195. [PMID: 34527169 PMCID: PMC8437592 DOI: 10.1155/2021/1377195] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 12/14/2022]
Abstract
Although hippocampal changes due to noise-induced hearing loss have been suggested, little is known about the miRNA levels due to these hippocampal changes. Three-week-old Sprague-Dawley rats were divided into noise and control groups (n = 20 per group). The noise group rats were exposed to white Gaussian noise (115 dB SPL, 4 hours per day) for three days. One day after noise exposure, the hippocampi of rats were harvested and miRNA expressions were analyzed using the Affymetrix miRNA 4.0 microarray (n = 6 per group). The predicted target genes of each miRNA were retrieved, and the pathways related to the predicted target genes were analyzed. miR-758-5p, miR-210-5p, miR-370-5p, miR-652-5p, miR-3544, miR-128-1-5p, miR-665, miR-188-5p, and miR-874-5p expression increased in the hippocampal tissue of the noise group compared to that in the control group. The overlapping predicted target genes included Bend4, Creb1, Adcy6, Creb5, Kcnj9, and Pten. The pathways related to these genes were the estrogen signaling pathway, vasopressin-regulated water reabsorption, thyroid hormone synthesis, aldosterone synthesis and secretion, insulin secretion, circadian entrainment, insulin resistance, cholinergic synapse, dopaminergic synapse, cGMP-PKG signaling pathway, cAMP signaling pathway, PI3K-Akt signaling pathway, TNF signaling pathway, and AMPK signaling pathway. miR-448-3p, miR204-5p, and miR-204-3p expression decreased in the hippocampal tissue of the noise group compared to that in the control group. The overlapping predicted target genes of these three miRNAs were Rps6kas, Nfactc3, Rictor, Spred1, Cdh4, Cdh6, Dvl3, and Rcyt1b. Pathway analysis suggested that the Wnt signaling pathway is related to Dvl3 and Nfactc3. Noise-induced hearing loss dysregulates miR-758-5p, miR210-5p, miR370-5p, miR-652-5p, miR-3544, miR-128-1-5p, miR-665, miR-188-5p, miR-874-5p, miR-448-3p, miR-204-5p, miR-204-3p, and miR-140-5p expression in the hippocampus. These miRNAs have been predicted to be associated with hormonal, inflammatory, and synaptic pathways.
Collapse
|
16
|
MiR-377-3p inhibits atherosclerosis-associated vascular smooth muscle cell proliferation and migration via targeting neuropilin2. Biosci Rep 2021; 40:223827. [PMID: 32373927 PMCID: PMC7295640 DOI: 10.1042/bsr20193425] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 04/12/2020] [Accepted: 04/21/2020] [Indexed: 12/22/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) proliferation and migration are vital to atherosclerosis (AS) development and plaque rupture. MicroRNA-377-3p (miR-377-3p) has been reported to inhibit AS in apolipoprotein E knockout (ApoE−/−) mice. Herein, the mechanism underlying the effect of miR-377-3p on alleviating AS is explored. In vivo experiments, ApoE−/− mice were fed with high-fat diet (HFD) to induce AS and treated with miR-377-3p agomir or negative control agomir (agomir-NC) on week 0, 2, 4, 6, 8, 10 after HFD feeding. MiR-377-3p was found to restore HFD-induced AS lesions and expressions of matrix metalloproteinase (MMP)-2, MMP-9, α-smooth muscle actin (α-actin) and calponin. In in vitro experiments, human VSMCs were tranfected with miR-377-3p agomir or agomir-NC, followed by treatment with oxidized low-density lipoprotein (ox-LDL). MiR-377-3p was observed to significantly inhibit ox-LDL-induced VSMC proliferation characterized by inhibited cell viability, expressions of proliferating cell nuclear antigen (PCNA), cyclin D1 and cyclin E and cell cycle transition from G1 to S phase accompanied with less 5-Ethynyl-2′-deoxyuridine (EdU)-positive cells. Furthermore, MiR-377-3p significantly inhibited ox-LDL-induced VSMC migration characterized by inhibited wound closure and decreased relative VSMC migration. Besides, neuropilin2 (NRP2) was verified as a target of miR-377-3p. MiR-377-3p was observed to inhibit NRP2 expressions in vivo and in vitro. Moreover, miR-377-3p significantly inhibited MMP-2 and MMP-9 expressions in human VSMCs. Additionally, miR-377-3p-induced inhibition of VSMC proliferation and migration could be attenuated by NRP2 overexpression. These results indicated that miR-377-3p inhibited VSMC proliferation and migration via targeting NRP2. The present study provides an underlying mechanism for miR-377-3p-based AS therapy.
Collapse
|
17
|
Siedlecki-Wullich D, Miñano-Molina AJ, Rodríguez-Álvarez J. microRNAs as Early Biomarkers of Alzheimer's Disease: A Synaptic Perspective. Cells 2021; 10:113. [PMID: 33435363 PMCID: PMC7827653 DOI: 10.3390/cells10010113] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Pathogenic processes underlying Alzheimer's disease (AD) affect synaptic function from initial asymptomatic stages, long time before the onset of cognitive decline and neurodegeneration. Therefore, reliable biomarkers enabling early AD diagnosis and prognosis are needed to maximize the time window for therapeutic interventions. MicroRNAs (miRNAs) have recently emerged as promising cost-effective and non-invasive biomarkers for AD, since they can be readily detected in different biofluids, including cerebrospinal fluid (CSF) and blood. Moreover, a growing body of evidence indicates that miRNAs regulate synaptic homeostasis and plasticity processes, suggesting that they may be involved in early synaptic dysfunction during AD. Here, we review the current literature supporting a role of miRNAs during early synaptic deficits in AD, including recent studies evaluating their potential as AD biomarkers. Besides targeting genes related to Aβ and tau metabolism, several miRNAs also regulate synaptic-related proteins and transcription factors implicated in early synaptic deficits during AD. Furthermore, individual miRNAs and molecular signatures have been found to distinguish between prodromal AD and healthy controls. Overall, these studies highlight the relevance of considering synaptic-related miRNAs as potential biomarkers of early AD stages. However, further validation studies in large cohorts, including longitudinal studies, as well as implementation of standardized protocols, are needed to establish miRNA-based biomarkers as reliable diagnostic and prognostic tools.
Collapse
Affiliation(s)
- Dolores Siedlecki-Wullich
- Department Bioquímica i Biologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (A.J.M.-M.); (J.R.-Á.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 528031 Madrid, Spain
| | - Alfredo J. Miñano-Molina
- Department Bioquímica i Biologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (A.J.M.-M.); (J.R.-Á.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 528031 Madrid, Spain
| | - José Rodríguez-Álvarez
- Department Bioquímica i Biologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain; (A.J.M.-M.); (J.R.-Á.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 528031 Madrid, Spain
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
18
|
Gu X, Zhu J. Roles of Exosomes and Exosomal MicroRNAs in Postoperative Sleep Disturbance. Nat Sci Sleep 2021; 13:1363-1375. [PMID: 34354381 PMCID: PMC8331078 DOI: 10.2147/nss.s310351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 07/14/2021] [Indexed: 12/30/2022] Open
Abstract
Postoperative sleep disturbance (PSD) often occurs in elderly patients after major surgery and exerts harmful effects on postoperative recovery. PSD may increase the incidence of postoperative fatigue, severe anxiety and depression, pain sensitivity, and cognitive dysfunction, which can cause or aggravate neurodegenerative diseases via amyloid aggregation and tau accumulation. Exosomes are important carriers that mediate the transfer of active substances and genetic information among cells. Recent evidence has shown that exosomes are involved in the pathogenesis of end-organ morbidity caused by sleep disorders via increasing amyloid plaque formation, transmitting tau protein, regulating neuroinflammation, and increasing blood-brain barrier permeability. Additionally, exosomes may be useful for delivering therapeutic genetic materials, such as microRNAs (miRNAs) and proteins, to exert neuroprotective effects and reduce cognitive impairment. However, the molecular mechanisms underlying this process remain to be fully elucidated. This review focuses on exosome-related pathways and the modulatory role of exosomal miRNAs on the pathogenesis of sleep disturbance and neurodegeneration. Moreover, we discuss the advantages of reducing neurotoxic proteins via exosomal intervention and miRNA regulation. Future research in exosome administration may offer new insights into PSD-related pathomechanisms and therapeutics.
Collapse
Affiliation(s)
- Xiangyi Gu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Junchao Zhu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| |
Collapse
|
19
|
Bame M, McInnis MG, O'Shea KS. MicroRNA Alterations in Induced Pluripotent Stem Cell-Derived Neurons from Bipolar Disorder Patients: Pathways Involved in Neuronal Differentiation, Axon Guidance, and Plasticity. Stem Cells Dev 2020; 29:1145-1159. [PMID: 32438891 PMCID: PMC7469698 DOI: 10.1089/scd.2020.0046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/21/2020] [Indexed: 12/17/2022] Open
Abstract
Bipolar disorder (BP) is a complex psychiatric condition characterized by severe fluctuations in mood for which underlying pathological mechanisms remain unclear. Family and twin studies have identified a hereditary component to the disorder, but a single causative gene (or set of genes) has not been identified. MicroRNAs (miRNAs) are small, noncoding RNAs ∼20 nucleotides in length, that are responsible for the posttranslational regulation of multiple genes. They have been shown to play important roles in neural development as well as in the adult brain, and several miRNAs have been reported to be dysregulated in postmortem brain tissue isolated from bipolar patients. Because there are no viable cellular models to study BP, we have taken advantage of the recent discovery that somatic cells can be reprogrammed to pluripotency then directed to form the full complement of neural cells. Analysis of RNAs extracted from Control and BP patient-derived neurons identified 58 miRNAs that were differentially expressed between the two groups. Using quantitative polymerase chain reaction we validated six miRNAs that were elevated and two miRNAs that were expressed at lower levels in BP-derived neurons. Analysis of the targets of the miRNAs indicate that they may regulate a number of cellular pathways, including axon guidance, Mapk, Ras, Hippo, Neurotrophin, and Wnt signaling. Many are involved in processes previously implicated in BP, such as cell migration, axon guidance, dendrite and synapse development, and function. We have validated targets of several different miRNAs, including AXIN2, BDNF, RELN, and ANK3 as direct targets of differentially expressed miRNAs using luciferase assays. Identification of pathways altered in patient-derived neurons suggests that disruption of these regulatory networks that may contribute to the complex phenotypes in BP.
Collapse
Affiliation(s)
- Monica Bame
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Melvin G. McInnis
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - K. Sue O'Shea
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
20
|
A Novel Mechanism Underlying Activity-Dependent Pruning in Postnatal Prefrontal Cortex. J Neurosci 2020; 40:2186-2188. [PMID: 32161181 DOI: 10.1523/jneurosci.2394-19.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/22/2020] [Accepted: 01/27/2020] [Indexed: 11/21/2022] Open
|
21
|
Zhao X, Ding J, Pan H, Zhang S, Pan D, Yu H, Ye Z, Hua T. Anodal and cathodal tDCS modulate neural activity and selectively affect GABA and glutamate syntheses in the visual cortex of cats. J Physiol 2020; 598:3727-3745. [PMID: 32506434 DOI: 10.1113/jp279340] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS The present study showed that anodal and cathodal transcranial direct current stimulation (tDCS) can respectively increase and decrease the amplitude of visually evoked field potentials in the stimulated visual cortex of cats, with the effect lasting for ∼60-70 min. We directly measured tDCS-induced changes in the concentration of inhibitory and excitatory neurotransmitters in the visual cortex using the enzyme-linked immunosorbent assay method and showed that anodal and cathodal tDCS can selectively decrease the concentration of GABA and glutamate in the stimulated cortical area. Anodal and cathodal tDCS can selectively inhibit the synthesis of GABA and glutamate by suppressing the expression of GABA- and glutamate-synthesizing enzymes, respectively. ABSTRACT Transcranial direct current stimulation (tDCS) evokes long-lasting neuronal excitability in the target brain region. The underlying neural mechanisms remain poorly understood. The present study examined tDCS-induced alterations in neuronal activities, as well as the concentration and synthesis of GABA and glutamate (GLU), in area 21a (A21a) of cat visual cortex. Our analysis showed that anodal and cathodal tDCS respectively enhanced and suppressed neuronal activities in A21a, as indicated by a significantly increased and decreased amplitude of visually evoked field potentials (VEPs). The tDCS-induced effect lasted for ∼60-70 min. By contrast, sham tDCS had no significant impact on the VEPs in A21a. On the other hand, the concentration of GABA, but not that of GLU, in A21a significantly decreased after anodal tDCS relative to sham tDCS, whereas the concentration of GLU, but not that of GABA, in A21a significantly decreased after cathodal tDCS relative to sham tDCS. Furthermore, the expression of GABA-synthesizing enzymes GAD65 and GAD67 in A21a significantly decreased in terms of both mRNA and protein concentrations after anodal tDCS relative to sham tDCS, whereas that of GLU-synthesizing enzyme glutaminase (GLS) did not change significantly after anodal tDCS. By contrast, both mRNA and protein concentrations of GLS in A21a significantly decreased after cathodal tDCS relative to sham tDCS, whereas those of GAD65/GAD67 showed no significant change after cathodal tDCS. Taken together, these results indicate that anodal and cathodal tDCS may selectively reduce GABA and GLU syntheses and thus respectively enhance and suppress neuronal excitability in the stimulated brain area.
Collapse
Affiliation(s)
- Xiaojing Zhao
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
| | - Jian Ding
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
| | - Huijun Pan
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
| | - Shen Zhang
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
| | - Deng Pan
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
| | - Hao Yu
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
| | - Zheng Ye
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
| | - Tianmiao Hua
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
| |
Collapse
|
22
|
miRNAs-dependent regulation of synapse formation and function. Genes Genomics 2020; 42:837-845. [DOI: 10.1007/s13258-020-00940-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022]
|
23
|
Reza-Zaldivar EE, Hernández-Sápiens MA, Minjarez B, Gómez-Pinedo U, Sánchez-González VJ, Márquez-Aguirre AL, Canales-Aguirre AA. Dendritic Spine and Synaptic Plasticity in Alzheimer's Disease: A Focus on MicroRNA. Front Cell Dev Biol 2020; 8:255. [PMID: 32432108 PMCID: PMC7214692 DOI: 10.3389/fcell.2020.00255] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 03/26/2020] [Indexed: 12/12/2022] Open
Abstract
Dendrites and dendritic spines are dynamic structures with pivotal roles in brain connectivity and have been recognized as the locus of long-term synaptic plasticity related to cognitive processes such as learning and memory. In neurodegenerative diseases, the spine dynamic morphology alteration, such as shape and spine density, affects functional characteristics leading to synaptic dysfunction and cognitive impairment. Recent evidence implicates dendritic spine dysfunction as a critical feature in the pathogenesis of dementia, particularly Alzheimer’s disease. The alteration of spine morphology and their loss is correlated with the cognitive decline in Alzheimer’s disease patients even in the absence of neuronal loss, however, the underlying mechanisms are poorly understood. Currently, the microRNAs have emerged as essential regulators of synaptic plasticity. The changes in neuronal microRNA expression that contribute to the modification of synaptic function through the modulation of dendritic spine morphology or by regulating the local protein translation to synaptic transmission are determinant for synapse formation and synaptic plasticity. Focusing on microRNA and its targets may provide insight into new therapeutic opportunities. In this review we summarize the experimental evidence of the role that the microRNA plays in dendritic spine remodeling and synaptic plasticity and its potential therapeutic approach in Alzheimer’s disease. Targeting synaptic deficits through the structural alteration of dendritic spines could form part of therapeutic strategies to improve synaptic plasticity and to ameliorate cognitive impairments in Alzheimer’s disease and other neurological diseases.
Collapse
Affiliation(s)
| | | | - Benito Minjarez
- University Center of Biological and Agricultural Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Ulises Gómez-Pinedo
- Institute of Neurosciences, IdISSC, San Carlos Clinical Hospital, Madrid, Spain
| | | | - Ana Laura Márquez-Aguirre
- Medical and Pharmaceutical Biotechnology Unit, CIATEJ, Guadalajara, Mexico.,Preclinical Evaluation Unit, CIATEJ, Guadalajara, Mexico
| | - Alejandro Arturo Canales-Aguirre
- Medical and Pharmaceutical Biotechnology Unit, CIATEJ, Guadalajara, Mexico.,Preclinical Evaluation Unit, CIATEJ, Guadalajara, Mexico
| |
Collapse
|
24
|
Daws SE, Jamieson S, de Nijs L, Jones M, Snijders C, Klengel T, Joseph NF, Krauskopf J, Kleinjans J, Vinkers CH, Boks MPM, Geuze E, Vermetten E, Berretta S, Ressler KJ, Rutten BPF, Rumbaugh G, Miller CA. MicroRNA regulation of persistent stress-enhanced memory. Mol Psychiatry 2020; 25:965-976. [PMID: 31142820 PMCID: PMC6883139 DOI: 10.1038/s41380-019-0432-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/04/2019] [Accepted: 04/11/2019] [Indexed: 12/28/2022]
Abstract
Disruption of persistent, stress-associated memories is relevant for treating posttraumatic stress disorder (PTSD) and related syndromes, which develop in a subset of individuals following a traumatic event. We previously developed a stress-enhanced fear learning (SEFL) paradigm in inbred mice that produces PTSD-like characteristics in a subset of mice, including persistently enhanced memory and heightened cFos in the basolateral amygdala complex (BLC) with retrieval of the remote (30-day-old) stress memory. Here, the contribution of BLC microRNAs (miRNAs) to stress-enhanced memory was investigated because of the molecular complexity they achieve through their ability to regulate multiple targets simultaneously. We performed small-RNA sequencing (smRNA-Seq) and quantitative proteomics on BLC tissue collected from mice 1 month after SEFL and identified persistently changed microRNAs, including mir-135b-5p, and proteins associated with PTSD-like heightened fear expression. Viral-mediated overexpression of mir-135b-5p in the BLC of stress-resilient animals enhanced remote fear memory expression and promoted spontaneous renewal 14 days after extinction. Conversely, inhibition of BLC mir-135b-5p in stress-susceptible animals had the opposite effect, promoting a resilient-like phenotype. mir-135b-5p is highly conserved across mammals and was detected in post mortem human amygdala, as well as human serum samples. The mir-135b passenger strand, mir-135b-3p, was significantly elevated in serum from PTSD military veterans, relative to combat-exposed control subjects. Thus, miR-135b-5p may be an important therapeutic target for dampening persistent, stress-enhanced memory and its passenger strand a potential biomarker for responsivity to a mir-135-based therapeutic.
Collapse
Affiliation(s)
- Stephanie E Daws
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Sarah Jamieson
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Laurence de Nijs
- School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
| | - Meghan Jones
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Clara Snijders
- School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
| | - Torsten Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Nadine F Joseph
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Julian Krauskopf
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands
| | - Jos Kleinjans
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands
| | - Christiaan H Vinkers
- Department of Psychiatry, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Department of Anatomy and Neurosciences, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
| | - Marco P M Boks
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Elbert Geuze
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
- Research Centre for Military Mental Healthcare, Ministry of Defence, Utrecht, The Netherlands
| | - Eric Vermetten
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
- Research Centre for Military Mental Healthcare, Ministry of Defence, Utrecht, The Netherlands
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
| | - Sabina Berretta
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Bart P F Rutten
- School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
| | - Gavin Rumbaugh
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Courtney A Miller
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA.
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA.
| |
Collapse
|
25
|
Zhang SF, Gao J, Liu CM. The Role of Non-Coding RNAs in Neurodevelopmental Disorders. Front Genet 2019; 10:1033. [PMID: 31824553 PMCID: PMC6882276 DOI: 10.3389/fgene.2019.01033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 09/25/2019] [Indexed: 12/24/2022] Open
Abstract
Non-coding RNAs, a group of ribonucleic acids that are ubiquitous in the body and do not encode proteins, emerge as important regulatory factors in almost all biological processes in the brain. Extensive studies have suggested the involvement of non-coding RNAs in brain development and neurodevelopmental disorders, and dysregulation of non-coding RNAs is associated with abnormal brain development and the etiology of neurodevelopmental disorders. Here we provide an overview of the roles and working mechanisms of non-coding RNAs, and discuss potential clinical applications of non-coding RNAs as diagnostic and prognostic markers and as therapeutic targets in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Shuang-Feng Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Jun Gao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medicine Sciences & Peking Union Medical College, Beijing, China
| | - Chang-Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
26
|
Horch HW, Spicer SB, Low IIC, Joncas CT, Quenzer ED, Okoya H, Ledwidge LM, Fisher HP. Characterization of plexinA and two distinct semaphorin1a transcripts in the developing and adult cricket Gryllus bimaculatus. J Comp Neurol 2019; 528:687-702. [PMID: 31621906 DOI: 10.1002/cne.24790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/26/2019] [Accepted: 09/26/2019] [Indexed: 11/06/2022]
Abstract
Guidance cues act during development to guide growth cones to their proper targets in both the central and peripheral nervous systems. Experiments in many species indicate that guidance molecules also play important roles after development, though less is understood about their functions in the adult. The Semaphorin family of guidance cues, signaling through Plexin receptors, influences the development of both axons and dendrites in invertebrates. Semaphorin functions have been extensively explored in Drosophila melanogaster and some other Dipteran species, but little is known about their function in hemimetabolous insects. Here, we characterize sema1a and plexA in the cricket Gryllus bimaculatus. In fact, we found two distinct predicted Sema1a proteins in this species, Sema1a.1 and Sema1a.2, which shared only 48% identity at the amino acid level. We include a phylogenetic analysis that predicted that many other insect species, both holometabolous and hemimetabolous, express two Sema1a proteins as well. Finally, we used in situ hybridization to show that sema1a.1 and sema1a.2 expression patterns were spatially distinct in the embryo, and both roughly overlap with plexA. All three transcripts were also expressed in the adult brain, mainly in the mushroom bodies, though sema1a.2 was expressed most robustly. sema1a.2 was also expressed strongly in the adult thoracic ganglia while sema1a.1 was only weakly expressed and plexA was undetectable.
Collapse
Affiliation(s)
- Hadley W Horch
- Department of Biology and Neuroscience, Bowdoin College, Brunswick, Maine
| | - Sara B Spicer
- Department of Biology and Neuroscience, Bowdoin College, Brunswick, Maine
| | - Isabel I C Low
- Department of Biology and Neuroscience, Bowdoin College, Brunswick, Maine
| | - Colby T Joncas
- Department of Biology and Neuroscience, Bowdoin College, Brunswick, Maine
| | - Eleanor D Quenzer
- Department of Biology and Neuroscience, Bowdoin College, Brunswick, Maine
| | - Hikmah Okoya
- Department of Biology and Neuroscience, Bowdoin College, Brunswick, Maine
| | - Lisa M Ledwidge
- Department of Biology and Neuroscience, Bowdoin College, Brunswick, Maine
| | - Harrison P Fisher
- Department of Biology and Neuroscience, Bowdoin College, Brunswick, Maine
| |
Collapse
|
27
|
Kiltschewskij D, Cairns MJ. Temporospatial guidance of activity-dependent gene expression by microRNA: mechanisms and functional implications for neural plasticity. Nucleic Acids Res 2019; 47:533-545. [PMID: 30535081 PMCID: PMC6344879 DOI: 10.1093/nar/gky1235] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/30/2018] [Indexed: 01/08/2023] Open
Abstract
MicroRNA are major regulators of neuronal gene expression at the post-transcriptional and translational levels. This layer of control is critical for spatially and temporally restricted gene expression, facilitating highly dynamic changes to cellular structure and function associated with neural plasticity. Investigation of microRNA function in the neural system, however, is at an early stage, and many aspects of the mechanisms employing these small non-coding RNAs remain unclear. In this article, we critically review current knowledge pertaining to microRNA function in neural activity, with emphasis on mechanisms of microRNA repression, their subcellular remodelling and functional impacts on neural plasticity and behavioural phenotypes.
Collapse
Affiliation(s)
- Dylan Kiltschewskij
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2323, Australia.,Centre for Brain and Mental Health Research, Hunter Medical Research Institute, New Lambton, NSW, 2323, Australia
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2323, Australia.,Centre for Brain and Mental Health Research, Hunter Medical Research Institute, New Lambton, NSW, 2323, Australia
| |
Collapse
|
28
|
Hegde AN, Smith SG. Recent developments in transcriptional and translational regulation underlying long-term synaptic plasticity and memory. ACTA ACUST UNITED AC 2019; 26:307-317. [PMID: 31416904 PMCID: PMC6699410 DOI: 10.1101/lm.048769.118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/20/2019] [Indexed: 12/16/2022]
Abstract
Formation of long-term synaptic plasticity that underlies long-term memory requires new protein synthesis. Years of research has elucidated some of the transcriptional and translational mechanisms that contribute to the production of new proteins. Early research on transcription focused on the transcription factor cAMP-responsive element binding protein. Since then, other transcription factors, such as the Nuclear Receptor 4 family of proteins that play a role in memory formation and maintenance have been identified. In addition, several studies have revealed details of epigenetic mechanisms consisting of new types of chemical alterations of DNA such as hydroxymethylation, and various histone modifications in long-term synaptic plasticity and memory. Our understanding of translational control critical for memory formation began with the identification of molecules that impinge on the 5′ and 3′ untranslated regions of mRNAs and continued with the appreciation for local translation near synaptic sites. Lately, a role for noncoding RNAs such as microRNAs in regulating translation factors and other molecules critical for memory has been found. This review describes the past research in brief and mainly focuses on the recent work on molecular mechanisms of transcriptional and translational regulation that form the underpinnings of long-term synaptic plasticity and memory.
Collapse
Affiliation(s)
- Ashok N Hegde
- Department of Biological and Environmental Sciences, Georgia College and State University, Milledgeville, Georgia 31061, USA
| | - Spencer G Smith
- Department of Biological and Environmental Sciences, Georgia College and State University, Milledgeville, Georgia 31061, USA
| |
Collapse
|
29
|
Li Z, Jagadapillai R, Gozal E, Barnes G. Deletion of Semaphorin 3F in Interneurons Is Associated with Decreased GABAergic Neurons, Autism-like Behavior, and Increased Oxidative Stress Cascades. Mol Neurobiol 2019; 56:5520-5538. [PMID: 30635860 PMCID: PMC6614133 DOI: 10.1007/s12035-018-1450-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/07/2018] [Indexed: 12/11/2022]
Abstract
Autism and epilepsy are diseases which have complex genetic inheritance. Genome-wide association and other genetic studies have implicated at least 500+ genes associated with the occurrence of autism spectrum disorders (ASD) including the human semaphorin 3F (Sema 3F) and neuropilin 2 (NRP2) genes. However, the genetic basis of the comorbid occurrence of autism and epilepsy is unknown. The aberrant development of GABAergic circuitry is a possible risk factor in autism and epilepsy. Molecular biological approaches were used to test the hypothesis that cell-specific genetic variation in mouse homologs affects the formation and function of GABAergic circuitry. The empirical analysis with mice homozygous null for one of these genes, Sema 3F, in GABAergic neurons substantiated these predictions. Notably, deletion of Sema 3F in interneurons but not excitatory neurons during early development decreased the number of interneurons/neurites and mRNAs for cell-specific GABAergic markers and increased epileptogenesis and autistic behaviors. Studies of interneuron cell-specific knockout of Sema 3F signaling suggest that deficient Sema 3F signaling may lead to neuroinflammation and oxidative stress. Further studies of mouse KO models of ASD genes such as Sema 3F or NRP2 may be informative to clinical phenotypes contributing to the pathogenesis in autism and epilepsy patients.
Collapse
Affiliation(s)
- Zhu Li
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Rekha Jagadapillai
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Evelyne Gozal
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Gregory Barnes
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA.
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, USA.
- Pediatric Research Institute, University of Louisville Autism Center, 1405 East Burnett Ave, Louisville, KY, 40217, USA.
| |
Collapse
|
30
|
Wang Q, Liu Z, Lin Z, Zhang R, Lu Y, Su W, Li F, Xu X, Tu M, Lou Y, Zhao J, Zheng X. De Novo Germline Mutations in SEMA5A Associated With Infantile Spasms. Front Genet 2019; 10:605. [PMID: 31354784 PMCID: PMC6635550 DOI: 10.3389/fgene.2019.00605] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 06/07/2019] [Indexed: 11/13/2022] Open
Abstract
Infantile spasm (IS) is an early-onset epileptic encephalopathy that usually presents with hypsarrhythmia on an electroencephalogram with developmental impairment or regression. In this study, whole-exome sequencing was performed to detect potential pathogenic de novo mutations, and finally we identified a novel damaging de novo mutation in SEMA5A and a compound heterozygous mutation in CLTCL1 in three sporadic trios with IS. The expression profiling of SEMA5A in the human brain showed that it was mainly highly expressed in the cerebral cortex, during the early brain development stage (8 to 9 post-conception weeks and 0 to 5 months after birth). In addition, we identified a close protein-protein interaction network between SEMA5A and candidate genes associated with epilepsy, autism spectrum disorder (ASD) or intellectual disability. Gene enrichment and function analysis demonstrated that genes interacting with SEMA5A were significantly enriched in several brain regions across early fetal development, including the cortex, cerebellum, striatum and thalamus (q < 0.05), and were involved in axonal, neuronal and synapse-associated processes. Furthermore, SEMA5A and its interacting genes were associated with ASD, epilepsy syndrome and developmental disorders of mental health. Our results provide insightful information indicating that SEMA5A may contribute to the development of the brain and is associated with IS. However, further genetic studies are still needed to evaluate the role of SEMA5A in IS to definitively establish the role of SEMA5A in this disorder.
Collapse
Affiliation(s)
- Qiongdan Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhenwei Liu
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zhongdong Lin
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ru Zhang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yutian Lu
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Weijue Su
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Feng Li
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xi Xu
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Mengyun Tu
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yongliang Lou
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, Zhejiang, China
| | - Junzhao Zhao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiaoqun Zheng
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, Zhejiang, China
| |
Collapse
|
31
|
Abstract
In the past few decades, the field of neuroepigenetics has investigated how the brain encodes information to form long-lasting memories that lead to stable changes in behaviour. Activity-dependent molecular mechanisms, including, but not limited to, histone modification, DNA methylation and nucleosome remodelling, dynamically regulate the gene expression required for memory formation. Recently, the field has begun to examine how a learning experience is integrated at the level of both chromatin structure and synaptic physiology. Here, we provide an overview of key established epigenetic mechanisms that are important for memory formation. We explore how epigenetic mechanisms give rise to stable alterations in neuronal function by modifying synaptic structure and function, and highlight studies that demonstrate how manipulating epigenetic mechanisms may push the boundaries of memory.
Collapse
Affiliation(s)
- Rianne R Campbell
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, Center for Addiction Neuroscience, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, Center for Addiction Neuroscience, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA.
| |
Collapse
|
32
|
Qian Q, Zhang J, He FP, Bao WX, Zheng TT, Zhou DM, Pan HY, Zhang H, Zhang XQ, He X, Sun BG, Luo BY, Chen C, Peng GP. Down-regulated expression of microRNA-338-5p contributes to neuropathology in Alzheimer's disease. FASEB J 2018; 33:4404-4417. [PMID: 30576233 DOI: 10.1096/fj.201801846r] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Alzheimer's disease (AD) is a leading cause of dementia. However, the mechanisms responsible for development of AD, especially for the sporadic variant, are still not clear. In our previous study, we discovered that a small noncoding RNA (miR-188-3p) targeting β-site amyloid precursor protein cleaving enzyme (BACE)-1, a key enzyme responsible for Aβ formation, plays an important role in the development of neuropathology in AD. In the present study, we identified that miR-338-5p, a new miRNA that also targets BACE1, contributes to AD neuropathology. We observed that expression of miR-338-5p was significantly down-regulated in the hippocampus of patients with AD and 5XFAD transgenic (TG) mice, an animal model of AD. Overexpression of miR-338-5p in the hippocampus of TG mice reduced BACE1 expression, Aβ formation, and neuroinflammation. Overexpression of miR-338-5p functionally prevented impairments in long-term synaptic plasticity, learning ability, and memory retention in TG mice. In addition, we provide evidence that down-regulated expression of miR-338-5p in AD is regulated through the NF-κB signaling pathway. Our results suggest that down-regulated expression of miR-338-5p plays an important role in the development of AD.-Qian, Q., Zhang, J., He, F.-P., Bao, W.-X., Zheng, T.-T., Zhou, D.-M., Pan, H.-Y., Zhang, H., Zhang, X.-Q., He, X., Sun, B.-G., Luo, B.-Y., Chen, C., Peng, G.-P. Down-regulated expression of microRNA-338-5p contributes to neuropathology in Alzheimer's disease.
Collapse
Affiliation(s)
- Qi Qian
- Department of Neurology, Center for Brain Sciences, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China; and
| | - Jian Zhang
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Fang-Ping He
- Department of Neurology, Center for Brain Sciences, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wang-Xiao Bao
- Department of Neurology, Center for Brain Sciences, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting-Ting Zheng
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China.,Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Dong-Ming Zhou
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong-Yu Pan
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Heng Zhang
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-Qin Zhang
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao He
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing-Gui Sun
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Ben-Yan Luo
- Department of Neurology, Center for Brain Sciences, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chu Chen
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Guo-Ping Peng
- Department of Neurology, Center for Brain Sciences, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
33
|
Ren Z, Yu J, Wu Z, Si W, Li X, Liu Y, Zhou J, Deng R, Chen D. MicroRNA-210-5p Contributes to Cognitive Impairment in Early Vascular Dementia Rat Model Through Targeting Snap25. Front Mol Neurosci 2018; 11:388. [PMID: 30483048 PMCID: PMC6243094 DOI: 10.3389/fnmol.2018.00388] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/04/2018] [Indexed: 12/13/2022] Open
Abstract
Vascular dementia (VD) is the most common form of dementia in elderly people. However, little is understood about the role of microRNAs (miRNAs) involved in cognitive impairment in early VD. Here, a VD model induced by chronic cerebral ischemia and fetal bovine serum (FBS)-free cell model that detects synapse formation was established to investigate the function of miRNAs in early VD. The microarray analysis and real-time reverse transcription polymerase chain reaction (RT-PCR) showed that miR-210-5p increased significantly in the hippocampus of rats with 4 weeks of ischemia. The VD model rats also displayed significant cognitive deficits and synaptic loss. The overexpression of miR-210-5p decreased the synaptic number in primary hippocampal neurons, whereas specific suppression of miR-210-5p resulted in the formation of more synapses. Additionally, intracerebroventricular (ICV) injection of miR-210-5p agomir to VD rats aggravated phenotypes of cognitive impairment and synaptic loss. These VD-induced phenotypes were effectively attenuated by miR-210-5p antagomir. Moreover, bioinformatic prediction revealed that synaptosomal-associated protein of 25 KDa (Snap25) mRNA is targeted by miR-210-5p. The miR-210-5p decreased the luciferase activities of 3’ untranslated region (3’UTR) of Snap25 mRNA. Mutation of predicted miR-210-5p binding sites in the 3’ UTR of Snap25 mRNA abolished the miR-210-5p-induced decrease in luciferase activity. Western blot and immunofluorescence staining confirmed that miR-210-5p targets Snap25. Finally, RT-quantitative PCR (qPCR) and immunofluorescence staining detected that miR-210-5p agomir downregulated Snap25 expression in the cornu ammonis1 (CA1) region of hippocampi in VD rats, whereas miR-210-5p antagomir upregulated Snap25 expression. Altogether, miR-210-5p contributes to cognitive impairment in chronic ischemia-induced VD model through the regulation of Snap25 expression, which potentially provides an opportunity to develop a new therapeutic strategy for VD.
Collapse
Affiliation(s)
- Zhenxing Ren
- Department of Anatomy, The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junlong Yu
- College of Basic Medicine, The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zimei Wu
- Department of Anatomy, The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenwen Si
- Department of Anatomy, The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xianqian Li
- Department of Anatomy, The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuqing Liu
- Department of Anatomy, The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianhong Zhou
- Department of Anatomy, The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rudong Deng
- Department of Anatomy, The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dongfeng Chen
- Department of Anatomy, The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
34
|
Fainstein N, Dan-Goor N, Ben-Hur T. Resident brain neural precursor cells develop age-dependent loss of therapeutic functions in Alzheimer's mice. Neurobiol Aging 2018; 72:40-52. [PMID: 30205359 DOI: 10.1016/j.neurobiolaging.2018.07.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 06/25/2018] [Accepted: 07/26/2018] [Indexed: 12/27/2022]
Abstract
There is vast knowledge on pathogenic mechanisms in Alzheimer's disease but very little on means by which the brain protects itself from disease. A major candidate in providing neuroprotection is the resident brain neural precursor/stem cell (NPC) pool. Transplanted NPCs possess powerful immune-modulatory and trophic properties in vivo and in vitro, underscoring the question whether resident brain NPCs have any role in regulating disease pathology in Alzheimer's disease, and particularly whether they fail to protect the brain from degeneration. To evaluate brain NPC function in relation to disease pathology, we first characterized the pathological properties of 5xFAD transgenic mouse model of Alzheimer's disease at different ages. We found that age 7 months is a critical time point of heavy amyloid deposition and gliosis but before neurodegeneration and a normal basal rate of NPC turnover in the subventricular zone (SVZ) of 5xFAD mice as compared to wild-type mice. Analysis of NPC functional properties showed that despite preserved rate of turnover, there was substantial SVZ NPC dysfunction as indicated by both ex vivo and in vivo assays. Freshly isolated NPCs from 7-month-old 5xFAD mice exhibited reduced expansion rate and diminished immune-modulatory and trophic properties. Moreover, there was slowed recovery of SVZ NPCs after cytosine-arabinoside insult and markedly reduced migratory response following a lysolecithin-induced lesion in the corpus callosum in vivo. Importantly, these functions were fully preserved in 2-month-old 5xFAD mice, a time point before Alzheimer's disease-specific pathological changes. There was reduced expression of key genes involved in NPC proliferative and migratory response in NPCs derived from 7-month-old 5xFAD mice. The dysfunctional properties and downregulation of gene expression were reversible in NPCs derived from 7-month-old 5xFAD mice following in vitro expansion and were reproduced in wild-type NPC by addition of amyloid beta peptide. Thus, there is age-dependent acquired NPC dysfunction, with loss of immune-modulatory and neurotrophic properties, which is induced by the pathological Alzheimer's brain environment at a critical time point before neurodegeneration. We suggest that failure of resident NPC to provide tissue support may be involved in promoting neurodegeneration.
Collapse
Affiliation(s)
- Nina Fainstein
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Nadav Dan-Goor
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Tamir Ben-Hur
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
35
|
Zhao L, Ni X, Zhao L, Zhang Y, Jin D, Yin W, Wang D, Zhang W. MiroRNA-188 Acts as Tumor Suppressor in Non-Small-Cell Lung Cancer by Targeting MAP3K3. Mol Pharm 2018. [PMID: 29528232 DOI: 10.1021/acs.molpharmaceut.8b00071] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the most prevalent form of lung cancer. MicroRNAs have been increasingly implicated in NSCLC and may serve as novel therapeutic targets to combat cancer. Here we investigated the functional implication of miR-188 in NSCLC. We first analyzed miR-188 expression in both NSCLC clinical samples and cancer cell lines. Next we investigated its role in A549 and H2126 cells with cell proliferation, migration, and apoptosis assays. To extend the in vitro study, we employed both xenograft model and LSL- K-ras G12D lung cancer model to examine the role of miR-188 in tumorigenesis. Last we tested MAP3K3 as miR-188 target in NSCLC model. MiR-188 expression was significantly downregulated at the NSCLC tumor sites and lung cancer cells. In vitro transfection of miR-188 reduced cell proliferation and migration potential and promoted cell apoptosis. In xenograft model, miR-188 inhibited tumor growth derived from cancer cells. Intranasal miR-188 administration reduced tumor formation in NSCLC animal model. MAP3K3 was validated as direct target of miR-188. Knocking down MAP3K3 in mice also inhibited tumorigenesis in LSL- K-ras G12D model. Our results demonstrate that miR-188 and its downstream target MAP3K3 could be a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Lili Zhao
- Department of Respiratory Medicine , Affiliated Hongqi Hospital of Mudanjiang Medical University , Mudanjiang 157011 , Heilongjiang , P. R. China
| | - Xin Ni
- Department of Respiratory Medicine , Affiliated Hongqi Hospital of Mudanjiang Medical University , Mudanjiang 157011 , Heilongjiang , P. R. China
| | - Linlin Zhao
- Department of Respiratory Medicine , Affiliated Hongqi Hospital of Mudanjiang Medical University , Mudanjiang 157011 , Heilongjiang , P. R. China
| | - Yao Zhang
- Department of Respiratory Medicine , Affiliated Hongqi Hospital of Mudanjiang Medical University , Mudanjiang 157011 , Heilongjiang , P. R. China
| | - Dan Jin
- Department of Ultrasound , Mudanjiang Women and Children's Hospital , Mudanjiang 157000 , Heilongjiang , P. R. China
| | - Wei Yin
- Department of Bone Surgery , Mudanjiang Forestry Hospital , Mudanjiang 157000 , Heilongjiang , P. R. China
| | - Dandan Wang
- Department of Respiratory Medicine , Affiliated Hongqi Hospital of Mudanjiang Medical University , Mudanjiang 157011 , Heilongjiang , P. R. China
| | - Wei Zhang
- Department of Respiratory Medicine , Affiliated Hongqi Hospital of Mudanjiang Medical University , Mudanjiang 157011 , Heilongjiang , P. R. China
| |
Collapse
|
36
|
Wang D, Wang X, Liu X, Jiang L, Yang G, Shi X, Zhang C, Piao F. Inhibition of miR-219 Alleviates Arsenic-Induced Learning and Memory Impairments and Synaptic Damage Through Up-regulating CaMKII in the Hippocampus. Neurochem Res 2018; 43:948-958. [DOI: 10.1007/s11064-018-2500-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 02/09/2018] [Accepted: 02/16/2018] [Indexed: 01/09/2023]
|
37
|
Mosakhani N, Sarhadi V, Panula P, Partinen M, Knuutila S. Narcolepsy patients' blood-based miRNA expression profiling: miRNA expression differences with Pandemrix vaccination. Acta Neurol Scand 2017; 136:462-469. [PMID: 28251619 DOI: 10.1111/ane.12749] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Narcolepsy is a neurological sleep disorder characterized by excessive daytime sleepiness and nighttime sleep disturbance. Among children and adolescents vaccinated with Pandemrix vaccine in Finland and Sweden, the number of narcolepsy cases increased. Our aim was to identify miRNAs involved in narcolepsy and their association with Pandemrix vaccination. MATERIALS AND METHODS We performed global miRNA proofing by miRNA microarrays followed by RT-PCR verification on 20 narcolepsy patients (Pandemrix-associated and Pandemrix-non-associated) and 17 controls (vaccinated and non-vaccinated). RESULTS Between all narcolepsy patients and controls, 11 miRNAs were differentially expressed; 17 miRNAs showed significantly differential expression between Pandemrix-non-associated narcolepsy patients and non-vaccinated healthy controls. MiR-188-5p and miR-4499 were over-expressed in narcolepsy patients vs healthy controls. Two miRNAs, miR-1470 and miR-4455, were under-expressed in Pandemrix-associated narcolepsy patients vs Pandemrix-non-associated narcolepsy patients. CONCLUSIONS We identified miRNA expression patterns in narcolepsy patients that linked them to mRNA targets known to be involved in brain-related pathways or brain disorders.
Collapse
Affiliation(s)
- N. Mosakhani
- Department of Pathology; University of Helsinki; Helsinki Finland
| | - V. Sarhadi
- Department of Pathology; University of Helsinki; Helsinki Finland
| | - P. Panula
- Neuroscience Center; Biomedicum; University of Helsinki; Helsinki Finland
| | - M. Partinen
- Department of Clinical Neurosciences; University of Helsinki; Helsinki Finland
- Helsinki Sleep Clinic; Vitalmed Research Center; Helsinki Finland
| | - S. Knuutila
- Department of Pathology; University of Helsinki; Helsinki Finland
| |
Collapse
|
38
|
Wei CW, Luo T, Zou SS, Wu AS. Research progress on the roles of microRNAs in governing synaptic plasticity, learning and memory. Life Sci 2017; 188:118-122. [PMID: 28866103 DOI: 10.1016/j.lfs.2017.08.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/24/2017] [Accepted: 08/25/2017] [Indexed: 12/16/2022]
Abstract
The importance of non-coding RNA involved in biological processes has become apparent in recent years and the mechanism of transcriptional regulation has also been identified. MicroRNAs (miRNAs) represent a class of small regulatory non-coding RNAs of 22bp in length that mediate gene silencing by identifying specific sequences in the target messenger RNAs (mRNAs). Many miRNAs are highly expressed in the central nervous system in a spatially and temporally controlled manner in normal physiology, as well as in certain pathological conditions. There is growing evidence that a considerable number of specific miRNAs play important roles in synaptic plasticity, learning and memory function. In addition, the dysfunction of these molecules may also contribute to the etiology of several neurodegenerative diseases. Here we provide an overview of the current literatures, which support non-coding RNA-mediated gene function regulation represents an important but underappreciated, layer of epigenetic control that facilitates learning and memory functions.
Collapse
Affiliation(s)
- Chang-Wei Wei
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Ting Luo
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Shan-Shan Zou
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - An-Shi Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
39
|
Ku T, Li B, Gao R, Zhang Y, Yan W, Ji X, Li G, Sang N. NF-κB-regulated microRNA-574-5p underlies synaptic and cognitive impairment in response to atmospheric PM 2.5 aspiration. Part Fibre Toxicol 2017; 14:34. [PMID: 28851397 PMCID: PMC5575838 DOI: 10.1186/s12989-017-0215-3] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 08/20/2017] [Indexed: 12/12/2022] Open
Abstract
Background PM2.5 (particulate matter ≤ 2.5 μm) is one of the leading environmental risk factors for the global burden of disease. Whereas increasing evidence has linked the adverse roles of PM2.5 with cardiovascular and respiratory diseases, limited but growing emerging evidence suggests that PM2.5 exposure can affect the nervous system, causing neuroinflammation, synaptic dysfunction and cognitive deterioration. However, the molecular mechanisms underlying the synaptic and cognitive deficits elicited by PM2.5 exposure are largely unknown. Methods C57BL/6 mice received oropharyngeal aspiration of PM2.5 (1 and 5 mg/kg bw) every other day for 4 weeks. The mice were also stereotaxically injected with β-site amyloid precursor protein cleaving enzyme 1 (β-secretase, BACE1) shRNA or LV-miR-574-5p lentiviral constructs in the absence or presence of PM2.5 aspiration at 5 mg/kg bw every other day for 4 weeks. Spatial learning and memory were assessed with the Morris water maze test, and synaptic function integrity was evaluated with electrophysiological recordings of long-term potentiation (LTP) and immunoblot analyses of glutamate receptor subunit expression. The expression of α-secretase (ADAM10), BACE1, and γ-secretase (nicastrin) and the synthesis and accumulation of amyloid β (Aβ) were measured by immunoblot and enzyme-linked immunosorbent assay (ELISA). MicroRNA (miRNA) expression was screened with a microRNA microarray analysis and confirmed by real-time quantitative reverse transcription PCR (qRT-PCR) analysis. Dual-luciferase reporter gene and chromatin immunoprecipitation (ChIP) analyses were used to detect the binding of miR-574-5p in the 3’UTR of BACE1 and NF-κB p65 in the promoter of miR-574-5p, respectively. Results PM2.5 aspiration caused neuroinflammation and deteriorated synaptic function integrity and spatial learning and memory, and the effects were associated with the induction of BACE1. The action was mediated by NF-κB p65-regulated downregulation of miR-574-5p, which targets BACE1. Overexpression of miR-574-5p in the hippocampal region decreased BACE1 expression, restored synaptic function, and improved spatial memory and learning following PM2.5 exposure. Conclusions Taken together, our findings reveal a novel molecular mechanism underlying impaired synaptic and cognitive function following exposure to PM2.5, suggesting that miR-574-5p is a potential intervention target for the prevention and treatment of PM2.5-induced neurological disorders. Electronic supplementary material The online version of this article (10.1186/s12989-017-0215-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tingting Ku
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Ben Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Rui Gao
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Yingying Zhang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Wei Yan
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Xiaotong Ji
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China.
| |
Collapse
|
40
|
Li Y, Xia H, Chen L, Zhang X. Sevoflurane induces endoplasmic reticulum stress mediated apoptosis inmouse hippocampal neuronal HT22 cells via modulating miR-15b-5p/Rab1A signaling pathway. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:8270-8280. [PMID: 31966678 PMCID: PMC6965408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 06/20/2017] [Indexed: 06/10/2023]
Abstract
Sevoflurane (Sev) is a widely used anaesthetic agent in clinical patients. Growing evidences indicated that Sev resulted in cognitive impairment via inducing endoplasmic reticulum (ER) stress mediated neurons apoptosis in vivo. However, the underlying molecular mechanisms have not yet fully understood. In this study, we found that Sev exposure suppresses cell viability, and induces apoptosis by activating caspase-3 apoptotic signaling pathway. Our results further verified that Sevtriggers ER stress via upregulating its markers glucose-regulated protein 78 (GRP78), C/EBP homologous protein (CHOP), caspase-12 and cleaved-PARP proteins. Recently, microRNAs (miRNAs) have been proven to regulate ER stress in a variety of cells, especially neuronal cells. Therefore, we performed the microarray analysis to identified miRNA levels in HT22 cells after treatment with Sev. Our results showed that Sev induces miRNAs aberrant expression and miR-15b-5p was one of the miRNAs being most upregulated in HT22 cells. Furthermore, the Sev-induced apoptosis and ER stress were rescued by knockdown of miR-15b-5p. Additionally, we demonstrated that miR-15b-5p suppresses Rab1A, a regulator in inducing ER stress, by directly targeting its 3'-UTR in HT22 cells. These results suggested that Sev exposure induces ER stress mediated apoptosis in HT22 cells via regulating miR-15b-5p/Rab1A signaling pathway. These data may provide an important therapeutic strategy for fighting against Sev through ER stress mediated neuronal apoptosis in clinical patients.
Collapse
Affiliation(s)
- Yongliang Li
- Department of Anesthesiology, The People's Hospital of Ruian (The Third Affiliated Hospital of Wenzhou Medical University) Ruian, Zhejiang, China
| | - Haijie Xia
- Department of Anesthesiology, The People's Hospital of Ruian (The Third Affiliated Hospital of Wenzhou Medical University) Ruian, Zhejiang, China
| | - Liangliang Chen
- Department of Anesthesiology, The People's Hospital of Ruian (The Third Affiliated Hospital of Wenzhou Medical University) Ruian, Zhejiang, China
| | - Xuzhong Zhang
- Department of Anesthesiology, The People's Hospital of Ruian (The Third Affiliated Hospital of Wenzhou Medical University) Ruian, Zhejiang, China
| |
Collapse
|
41
|
Kos A, Klein-Gunnewiek T, Meinhardt J, Loohuis NFMO, van Bokhoven H, Kaplan BB, Martens GJ, Kolk SM, Aschrafi A. MicroRNA-338 Attenuates Cortical Neuronal Outgrowth by Modulating the Expression of Axon Guidance Genes. Mol Neurobiol 2017; 54:3439-3452. [PMID: 27180071 PMCID: PMC5658782 DOI: 10.1007/s12035-016-9925-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/03/2016] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRs) are small non-coding RNAs that confer robustness to gene networks through post-transcriptional gene regulation. Previously, we identified miR-338 as a modulator of axonal outgrowth in sympathetic neurons. In the current study, we examined the role of miR-338 in the development of cortical neurons and uncovered its downstream mRNA targets. Long-term inhibition of miR-338 during neuronal differentiation resulted in reduced dendritic complexity and altered dendritic spine morphology. Furthermore, monitoring axon outgrowth in cortical cells revealed that miR-338 overexpression decreased, whereas inhibition of miR-338 increased axonal length. To identify gene targets mediating the observed phenotype, we inhibited miR-338 in cortical neurons and performed whole-transcriptome analysis. Pathway analysis revealed that miR-338 modulates a subset of transcripts involved in the axonal guidance machinery by means of direct and indirect gene targeting. Collectively, our results implicate miR-338 as a novel regulator of cortical neuronal maturation by fine-tuning the expression of gene networks governing cortical outgrowth.
Collapse
Affiliation(s)
- Aron Kos
- Department of Cognitive Neuroscience, Radboud university medical center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Teun Klein-Gunnewiek
- Department of Cognitive Neuroscience, Radboud university medical center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Julia Meinhardt
- Department of Cognitive Neuroscience, Radboud university medical center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Nikkie F M Olde Loohuis
- Department of Cognitive Neuroscience, Radboud university medical center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Hans van Bokhoven
- Department of Cognitive Neuroscience, Radboud university medical center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud university medical center, 6500 HB, Nijmegen, The Netherlands
| | - Barry B Kaplan
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Gerard J Martens
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
- Department of Molecular Animal Physiology, Radboud University, Nijmegen, The Netherlands
| | - Sharon M Kolk
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
- Department of Molecular Animal Physiology, Radboud University, Nijmegen, The Netherlands
| | - Armaz Aschrafi
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands.
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892, USA.
| |
Collapse
|
42
|
Hu Z, Li Z. miRNAs in synapse development and synaptic plasticity. Curr Opin Neurobiol 2017; 45:24-31. [PMID: 28334640 DOI: 10.1016/j.conb.2017.02.014] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 02/23/2017] [Accepted: 02/26/2017] [Indexed: 01/21/2023]
Abstract
Synapses are functional units of the nervous system, through which information is transferred between neurons. The development and activity-dependent modification of synapses require temporally and spatially controlled modulation of gene expression. microRNAs (miRNAs) have emerged as essential regulators of gene expression. They are small non-coding RNAs that regulate mRNA stability and translation by interacting with the 3' untranslated region (3' UTR) of mRNAs. miRNAs are located to neuronal processes to regulate protein synthesis locally and their expression is regulated by synaptic activity. This article reviews recent findings on the role of miRNAs in synapse development and synaptic plasticity.
Collapse
Affiliation(s)
- Zhonghua Hu
- Section on Synapse Development and Plasticity, National Institute of Mental Health, National Institutes of Health, United States; Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, United States
| | - Zheng Li
- Section on Synapse Development and Plasticity, National Institute of Mental Health, National Institutes of Health, United States.
| |
Collapse
|
43
|
Millan MJ. Linking deregulation of non-coding RNA to the core pathophysiology of Alzheimer's disease: An integrative review. Prog Neurobiol 2017; 156:1-68. [PMID: 28322921 DOI: 10.1016/j.pneurobio.2017.03.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/09/2017] [Accepted: 03/09/2017] [Indexed: 02/06/2023]
Abstract
The human genome encodes a vast repertoire of protein non-coding RNAs (ncRNA), some specific to the brain. MicroRNAs, which interfere with the translation of target mRNAs, are of particular interest since their deregulation has been implicated in neurodegenerative disorders like Alzheimer's disease (AD). However, it remains challenging to link the complex body of observations on miRNAs and AD into a coherent framework. Using extensive graphical support, this article discusses how a diverse panoply of miRNAs convergently and divergently impact (and are impacted by) core pathophysiological processes underlying AD: neuroinflammation and oxidative stress; aberrant generation of β-amyloid-42 (Aβ42); anomalies in the production, cleavage and post-translational marking of Tau; impaired clearance of Aβ42 and Tau; perturbation of axonal organisation; disruption of synaptic plasticity; endoplasmic reticulum stress and the unfolded protein response; mitochondrial dysfunction; aberrant induction of cell cycle re-entry; and apoptotic loss of neurons. Intriguingly, some classes of miRNA provoke these cellular anomalies, whereas others act in a counter-regulatory, protective mode. Moreover, changes in levels of certain species of miRNA are a consequence of the above-mentioned anomalies. In addition to miRNAs, circular RNAs, piRNAs, long non-coding RNAs and other types of ncRNA are being increasingly implicated in AD. Overall, a complex mesh of deregulated and multi-tasking ncRNAs reciprocally interacts with core pathophysiological mechanisms underlying AD. Alterations in ncRNAs can be detected in CSF and the circulation as well as the brain and are showing promise as biomarkers, with the ultimate goal clinical exploitation as targets for novel modes of symptomatic and course-altering therapy.
Collapse
Affiliation(s)
- Mark J Millan
- Centre for Therapeutic Innovation in Neuropsychiatry, institut de recherche Servier, 125 chemin de ronde, 78290 Croissy sur Seine, France.
| |
Collapse
|
44
|
Ryan B, Logan BJ, Abraham WC, Williams JM. MicroRNAs, miR-23a-3p and miR-151-3p, Are Regulated in Dentate Gyrus Neuropil following Induction of Long-Term Potentiation In Vivo. PLoS One 2017; 12:e0170407. [PMID: 28125614 PMCID: PMC5268419 DOI: 10.1371/journal.pone.0170407] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/04/2017] [Indexed: 01/04/2023] Open
Abstract
Translation of synaptic mRNA contributes to alterations in the proteome necessary to consolidate long-term potentiation (LTP), a model of memory processes. Yet, how this process is controlled is not fully resolved. MicroRNAs are non-coding RNAs that negatively regulate gene expression by suppressing translation or promoting mRNA degradation. As specific microRNAs are synaptically located, we hypothesized that they are ideally suited to couple synaptic activation, translational regulation, and LTP persistence. The aim of this study was to identify LTP-regulated microRNAs at or near synapses. Accordingly, LTP was induced unilaterally at perforant path-dentate gyrus synapses in awake adult Sprague-Dawley rats. Five hours later, dentate gyrus middle molecular layer neuropil, containing potentiated synapses, was laser-microdissected. MicroRNA expression profiling, using TaqMan Low Density MicroRNA Microarrays (n = 4), identified eight regulated microRNAs. Subsequent individual TaqMan assays confirmed upregulation of miR-23a-3p (1.30 ± 0.10; p = 0.015) and miR-151-3p (1.17 ± 0.19; p = 0.045) in a second cohort (n = 7). Interestingly, bioinformatic analysis indicated that miR-151-3p and miR-23a-3p regulate synaptic reorganisation and transcription, respectively. In summary, we have demonstrated for the first time that microRNAs are regulated in isolated neuropil following LTP induction in vivo, supporting the hypothesis that synaptic, LTP-responsive microRNAs contribute to LTP persistence via regulation of the synaptic proteome.
Collapse
Affiliation(s)
- Brigid Ryan
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- The Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Barbara J. Logan
- The Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Wickliffe C. Abraham
- The Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Joanna M. Williams
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- The Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
- * E-mail:
| |
Collapse
|
45
|
Malan-Müller S, Hemmings S. The Big Role of Small RNAs in Anxiety and Stress-Related Disorders. ANXIETY 2017; 103:85-129. [DOI: 10.1016/bs.vh.2016.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
46
|
Song WY, Meng H, Wang XG, Jin HX, Yao GD, Shi SL, Wu L, Zhang XY, Sun YP. Reduced microRNA-188-3p expression contributes to apoptosis of spermatogenic cells in patients with azoospermia. Cell Prolif 2016; 50. [PMID: 27868267 DOI: 10.1111/cpr.12297] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 08/24/2016] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND AIMS Human mutL homologl (MLH1) works coordinately in sequential steps to initiate repair of DNA mismatches, and aberrant MLH1 expression is related to spermatogenetic malfunction. In the present study, MLH1 expression in patients with azoospermia was investigated, and moderating effects of miR-188-3p on MLH1 expression and spermatogenesis were identified. METHODS Testicular tissues from 16 patients with obstructive azoospermia (OA) and non-obstructive azoospermia (NOA), and tissues of eight healthy patients were collected. Real-time PCR, Western blotting and immunohistochemical staining were used to detect MLH1 expression. Chromatin immunoprecipitation assay and luciferase reporter assay were performed to evaluate histone acetylation level of miR-188-3p and relationships between miR-188-3p and MLH1. RESULTS Testicular MLH1 expression at mRNA and protein levels was significantly increased, while miR-188-3p expression was lower in patients with OA and NOA than that in controls. Reduced histone acetylation level of miR-188-3p promoter was observed in patients with azoospermia. Overexpression/inhibition of HDAC1, but not HDAC2, contributed to the significant reduction/increase of miR-188-3p expression. miR-188-3p targeted 3' UTR of MLH1 and regulated MLH1 expression. miR-188-3p inhibitor led to elevation of apoptotic level of spermatogenic cells in mice, while this effect was reversed by si-MLH1. CONCLUSION Down-regulation of miR-188-3p by reducing histone acetylation up-regulated MLH1 expression and contributed to promotion of apoptosis in spermatogenic cells, in patients with azoospermia.
Collapse
Affiliation(s)
- Wen-Yan Song
- Reproductive Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Meng
- Pathology Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xue-Gai Wang
- Reproductive Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hai-Xia Jin
- Reproductive Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gui-Dong Yao
- Reproductive Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Sen-Lin Shi
- Reproductive Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liang Wu
- Reproductive Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiang-Yang Zhang
- Reproductive Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying-Pu Sun
- Reproductive Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
47
|
Teppen TL, Krishnan HR, Zhang H, Sakharkar AJ, Pandey SC. The Potential Role of Amygdaloid MicroRNA-494 in Alcohol-Induced Anxiolysis. Biol Psychiatry 2016; 80:711-719. [PMID: 26786313 PMCID: PMC4882267 DOI: 10.1016/j.biopsych.2015.10.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 10/13/2015] [Accepted: 10/30/2015] [Indexed: 01/24/2023]
Abstract
BACKGROUND The antianxiety effects of ethanol appear to be a crucial factor in promoting alcohol intake. Regulation of gene expression by microRNA (miRNA) is an important epigenetic mechanism that affects neuronal pathways and behaviors. We investigated the role of miRNAs underlying the mechanisms of ethanol-induced anxiolysis. METHODS Acute ethanol-induced anxiolysis was measured in adult rats, and amygdaloid tissues were used for miRNA profiling by microarray analysis. The expression of miR-494 and its target genes in the amygdala was measured using real-time quantitative polymerase chain reaction. The direct role of miR-494 in the anxiety phenotype was also investigated via infusion of a miR-494 antagomir into the central nucleus of amygdala. RESULTS Microarray profiling of miRNAs in the amygdala showed significant alteration of several miRNA expression levels by acute ethanol exposure. Expression of miR-494 was significantly decreased, whereas expression of the binding protein of cyclic adenosine monophosphate response element binding protein (CBP), p300, and Cbp/p300-interacting transactivator 2 (Cited2) was increased in the amygdala during ethanol-induced anxiolysis. Inhibition of miR-494 in the central nucleus of amygdala, through infusion of a specific antagomir, provoked anxiolysis, mimicking the action of ethanol. Also, expression of Cited2, CBP, and p300 as well as histone H3-lysine 9 acetylation was significantly increased by miR-494 antagomir infusion, indicating their regulation by miR-494 in the amygdala. CONCLUSIONS These novel results suggest that acute ethanol-induced reduction in miR-494 expression in the amygdala can serve as a key regulatory mechanism for chromatin remodeling possibly leading to anxiolysis.
Collapse
Affiliation(s)
- Tara L. Teppen
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago IL 60612,Jesse Brown Veterans Affairs Medical Center Chicago, IL 60612
| | - Harish R. Krishnan
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago IL 60612,Jesse Brown Veterans Affairs Medical Center Chicago, IL 60612
| | - Huaibo Zhang
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago IL 60612,Jesse Brown Veterans Affairs Medical Center Chicago, IL 60612
| | - Amul J. Sakharkar
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago IL 60612,Jesse Brown Veterans Affairs Medical Center Chicago, IL 60612
| | - Subhash C. Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago IL 60612,Anatomy and Cell Biology, University of Illinois at Chicago, Chicago IL 60612,Jesse Brown Veterans Affairs Medical Center Chicago, IL 60612
| |
Collapse
|
48
|
Constantin L. The Role of MicroRNAs in Cerebellar Development and Autism Spectrum Disorder During Embryogenesis. Mol Neurobiol 2016; 54:6944-6959. [PMID: 27774573 DOI: 10.1007/s12035-016-0220-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/12/2016] [Indexed: 02/03/2023]
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNA molecules with wide-ranging and subtle effects on protein production. Their activity during the development of the cerebellum provides a valuable exemplar of how non-coding molecules may assist the development and function of the central nervous system and drive neurodevelopmental disorders. Three distinct aspects of miRNA contribution to early cerebellar development will here be reviewed. Aspects are the establishment of the cerebellar anlage, the generation and maturation of at least two principal cell types of the developing cerebellar microcircuit, and the etiology and early progression of autism spectrum disorder. It will be argued here that the autism spectrum is an adept model to explore miRNA impact on the cognitive and affective processes that descend from the developing cerebellum.
Collapse
Affiliation(s)
- Lena Constantin
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, 4072, Australia. .,Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
49
|
Lee K, Kim H, An K, Kwon OB, Park S, Cha JH, Kim MH, Lee Y, Kim JH, Cho K, Kim HS. Replenishment of microRNA-188-5p restores the synaptic and cognitive deficits in 5XFAD Mouse Model of Alzheimer's Disease. Sci Rep 2016; 6:34433. [PMID: 27708404 PMCID: PMC5052619 DOI: 10.1038/srep34433] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/12/2016] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs have emerged as key factors in development, neurogenesis and synaptic functions in the central nervous system. In the present study, we investigated a pathophysiological significance of microRNA-188-5p (miR-188-5p) in Alzheimer's disease (AD). We found that oligomeric Aβ1-42 treatment diminished miR-188-5p expression in primary hippocampal neuron cultures and that miR-188-5p rescued the Aβ1-42-mediated synapse elimination and synaptic dysfunctions. Moreover, the impairments in cognitive function and synaptic transmission observed in 7-month-old five familial AD (5XFAD) transgenic mice, were ameliorated via viral-mediated expression of miR-188-5p. miR-188-5p expression was down-regulated in the brain tissues from AD patients and 5XFAD mice. The addition of miR-188-5p rescued the reduction in dendritic spine density in the primary hippocampal neurons treated with oligomeric Aβ1-42 and cultured from 5XFAD mice. The reduction in the frequency of mEPSCs was also restored by addition of miR-188-5p. The impairments in basal fEPSPs and cognition observed in 7-month-old 5XFAD mice were ameliorated via the viral-mediated expression of miR-188-5p in the hippocampus. Furthermore, we found that miR-188 expression is CREB-dependent. Taken together, our results suggest that dysregulation of miR-188-5p expression contributes to the pathogenesis of AD by inducing synaptic dysfunction and cognitive deficits associated with Aβ-mediated pathophysiology in the disease.
Collapse
Affiliation(s)
- Kihwan Lee
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul, 110-799, Republic of Korea
| | - Hyunju Kim
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul, 110-799, Republic of Korea
| | - Kyongman An
- Department of Life Science, POSTECH, Pohang, Gyeongbuk, 790-784, Republic of Korea
| | - Oh-Bin Kwon
- Department of Life Science, POSTECH, Pohang, Gyeongbuk, 790-784, Republic of Korea
| | - Sungjun Park
- Department of Life Science, POSTECH, Pohang, Gyeongbuk, 790-784, Republic of Korea
| | - Jin Hee Cha
- Department of Physiology, College of Medicine, Seoul National University, Seoul, 110-799, Republic of Korea
| | - Myoung-Hwan Kim
- Department of Physiology, College of Medicine, Seoul National University, Seoul, 110-799, Republic of Korea
| | - Yoontae Lee
- Department of Life Science, POSTECH, Pohang, Gyeongbuk, 790-784, Republic of Korea
| | - Joung-Hun Kim
- Department of Life Science, POSTECH, Pohang, Gyeongbuk, 790-784, Republic of Korea
| | - Kwangwook Cho
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology (LINE), Faculty of Medicine and Dentistry, University of Bristol, Whitson Street, Bristol BS1 3NY, UK.,Centre for Synaptic Plasticity, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| | - Hye-Sun Kim
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul, 110-799, Republic of Korea.,Seoul National University Bundang Hospital, College of Medicine, Seoul National University, Seongnam, Gyeonggi, 463-707, Republic of Korea
| |
Collapse
|
50
|
Abstract
The last decade has been marked by an increased interest in relating epigenetic mechanisms to complex human behaviors, although this interest has not been balanced, accentuating various types of affective and primarily ignoring cognitive functioning. Recent animal model data support the view that epigenetic processes play a role in learning and memory consolidation and help transmit acquired memories even across generations. In this review, we provide an overview of various types of epigenetic mechanisms in the brain (DNA methylation, histone modification, and noncoding RNA action) and discuss their impact proximally on gene transcription, protein synthesis, and synaptic plasticity and distally on learning, memory, and other cognitive functions. Of particular importance are observations that neuronal activation regulates the dynamics of the epigenome's functioning under precise timing, with subsequent alterations in the gene expression profile. In turn, epigenetic regulation impacts neuronal action, closing the circle and substantiating the signaling pathways that underlie, at least partially, learning, memory, and other cognitive processes.
Collapse
|