1
|
Meccariello R, Bellenchi GC, Pulcrano S, D’Addario SL, Tafuri D, Mercuri NB, Guatteo E. Neuronal dysfunction and gene modulation by non-coding RNA in Parkinson's disease and synucleinopathies. Front Cell Neurosci 2024; 17:1328269. [PMID: 38249528 PMCID: PMC10796818 DOI: 10.3389/fncel.2023.1328269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/07/2023] [Indexed: 01/23/2024] Open
Abstract
Over the last few decades, emerging evidence suggests that non-coding RNAs (ncRNAs) including long-non-coding RNA (lncRNA), microRNA (miRNA) and circular-RNA (circRNA) contribute to the molecular events underlying progressive neuronal degeneration, and a plethora of ncRNAs have been identified significantly misregulated in many neurodegenerative diseases, including Parkinson's disease and synucleinopathy. Although a direct link between neuropathology and causative candidates has not been clearly established in many cases, the contribution of ncRNAs to the molecular processes leading to cellular dysfunction observed in neurodegenerative diseases has been addressed, suggesting that they may play a role in the pathophysiology of these diseases. Aim of the present Review is to overview and discuss recent literature focused on the role of RNA-based mechanisms involved in different aspects of neuronal pathology in Parkinson's disease and synucleinopathy models.
Collapse
Affiliation(s)
- Rosaria Meccariello
- Department of Medical and Movement Sciences and Wellness, University of Naples Parthenope, Naples, Italy
| | - Gian Carlo Bellenchi
- Institute of Genetics and Biophysics, CNR, Naples, Italy
- Experimental Neurology Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy
| | | | - Sebastian Luca D’Addario
- Experimental Neurology Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, CNR, Rome, Italy
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Domenico Tafuri
- Department of Medical and Movement Sciences and Wellness, University of Naples Parthenope, Naples, Italy
| | - Nicola B. Mercuri
- Experimental Neurology Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ezia Guatteo
- Department of Medical and Movement Sciences and Wellness, University of Naples Parthenope, Naples, Italy
- Experimental Neurology Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| |
Collapse
|
2
|
Zhou LT, Liu D, Kang HC, Lu L, Huang HZ, Ai WQ, Zhou Y, Deng MF, Li H, Liu ZQ, Zhang WF, Hu YZ, Han ZT, Zhang HH, Jia JJ, Sarkar AK, Sharaydeh S, Wang J, Man HY, Schilling M, Bertram L, Lu Y, Guo Z, Zhu LQ. Tau pathology epigenetically remodels the neuron-glial cross-talk in Alzheimer's disease. SCIENCE ADVANCES 2023; 9:eabq7105. [PMID: 37083538 PMCID: PMC10121173 DOI: 10.1126/sciadv.abq7105] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 03/22/2023] [Indexed: 05/03/2023]
Abstract
The neuron-glia cross-talk is critical to brain homeostasis and is particularly affected by neurodegenerative diseases. How neurons manipulate the neuron-astrocyte interaction under pathological conditions, such as hyperphosphorylated tau, a pathological hallmark in Alzheimer's disease (AD), remains elusive. In this study, we identified excessively elevated neuronal expression of adenosine receptor 1 (Adora1 or A1R) in 3×Tg mice, MAPT P301L (rTg4510) mice, patients with AD, and patient-derived neurons. The up-regulation of A1R was found to be tau pathology dependent and posttranscriptionally regulated by Mef2c via miR-133a-3p. Rebuilding the miR-133a-3p/A1R signal effectively rescued synaptic and memory impairments in AD mice. Furthermore, neuronal A1R promoted the release of lipocalin 2 (Lcn2) and resulted in astrocyte activation. Last, silencing neuronal Lcn2 in AD mice ameliorated astrocyte activation and restored synaptic plasticity and learning/memory. Our findings reveal that the tau pathology remodels neuron-glial cross-talk and promotes neurodegenerative progression. Approaches targeting A1R and modulating this signaling pathway might be a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Lan-Ting Zhou
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, Hubei 441053, China
| | - Dan Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hui-Cong Kang
- Department of Neurology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Lu Lu
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45229, USA
| | - He-Zhou Huang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wen-Qing Ai
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yang Zhou
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Man-Fei Deng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hao Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhi-Qiang Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wei-Feng Zhang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ya-Zhuo Hu
- Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Disease, Institute of Geriatrics, Chinese PLA General Hospital and Chinese PLA Medical Academy, Beijing, China
| | - Zhi-Tao Han
- Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Disease, Institute of Geriatrics, Chinese PLA General Hospital and Chinese PLA Medical Academy, Beijing, China
| | - Hong-Hong Zhang
- Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Disease, Institute of Geriatrics, Chinese PLA General Hospital and Chinese PLA Medical Academy, Beijing, China
| | - Jian-Jun Jia
- Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Disease, Institute of Geriatrics, Chinese PLA General Hospital and Chinese PLA Medical Academy, Beijing, China
| | - Avijite Kumer Sarkar
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45229, USA
| | - Saldin Sharaydeh
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45229, USA
| | - Jie Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, Hubei 430030, China
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Marcel Schilling
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Lübeck 23562, Germany
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Lübeck 23562, Germany
| | - Youming Lu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ziyuan Guo
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45229, USA
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
3
|
Kim GU, Sung SE, Kang KK, Choi JH, Lee S, Sung M, Yang SY, Kim SK, Kim YI, Lim JH, Seo MS, Lee GW. Therapeutic Potential of Mesenchymal Stem Cells (MSCs) and MSC-Derived Extracellular Vesicles for the Treatment of Spinal Cord Injury. Int J Mol Sci 2021; 22:13672. [PMID: 34948463 PMCID: PMC8703906 DOI: 10.3390/ijms222413672] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/14/2021] [Accepted: 12/18/2021] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) is a life-threatening condition that leads to permanent disability with partial or complete loss of motor, sensory, and autonomic functions. SCI is usually caused by initial mechanical insult, followed by a cascade of several neuroinflammation and structural changes. For ameliorating the neuroinflammatory cascades, MSC has been regarded as a therapeutic agent. The animal SCI research has demonstrated that MSC can be a valuable therapeutic agent with several growth factors and cytokines that may induce anti-inflammatory and regenerative effects. However, the therapeutic efficacy of MSCs in animal SCI models is inconsistent, and the optimal method of MSCs remains debatable. Moreover, there are several limitations to developing these therapeutic agents for humans. Therefore, identifying novel agents for regenerative medicine is necessary. Extracellular vesicles are a novel source for regenerative medicine; they possess nucleic acids, functional proteins, and bioactive lipids and perform various functions, including damaged tissue repair, immune response regulation, and reduction of inflammation. MSC-derived exosomes have advantages over MSCs, including small dimensions, low immunogenicity, and no need for additional procedures for culture expansion or delivery. Certain studies have demonstrated that MSC-derived extracellular vesicles (EVs), including exosomes, exhibit outstanding chondroprotective and anti-inflammatory effects. Therefore, we reviewed the principles and patho-mechanisms and summarized the research outcomes of MSCs and MSC-derived EVs for SCI, reported to date.
Collapse
Affiliation(s)
- Gang-Un Kim
- Department of Orthopedic Surgery, Hanil General Hospital, 308 Uicheon-ro, Dobong-gu, Seoul 01450, Korea;
| | - Soo-Eun Sung
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Kyung-Ku Kang
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Joo-Hee Choi
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Sijoon Lee
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Minkyoung Sung
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Seung Yun Yang
- Department of Biomaterials Science, Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea;
| | - Seul-Ki Kim
- Efficacy Evaluation Team, Food Science R&D Center, KolmarBNH CO., LTD, 61Heolleungro 8-gil, Seocho-gu, Seoul 06800, Korea;
| | | | - Ju-Hyeon Lim
- New Drug Development Center, Osong Medical Innovation Foundation, Chungbuk 28160, Korea;
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea
| | - Min-Soo Seo
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Gun Woo Lee
- Cellexobio, Co. Ltd., Daegu 42415, Korea;
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea
| |
Collapse
|
4
|
Potential of different cells-derived exosomal microRNA cargos for treating spinal cord injury. J Orthop Translat 2021; 31:33-40. [PMID: 34760623 PMCID: PMC8560648 DOI: 10.1016/j.jot.2021.09.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI) is a disastrous situation that affects many patients worldwide. A profound understanding of the pathology and etiology of SCI is of great importance in inspiring new therapeutic concepts and treatment. In recent years, exosomes, which are complex lipid membrane structures secreted nearly by all kinds of plants and animal cells, can transport their valuable cargoes (e.g., proteins, lipids, RNAs) to the targeted cells and exert their communication and regulation functions, which open up a new field of treatment of SCI. Notably, the exosome's advantage is transporting the carried material to the target cells across the blood-brain barrier and exerting regulatory functions. Among the cargoes of exosomes, microRNAs, through the modulation of their mRNA targets, emerges with great potentiality in the pathological process, diagnosis and treatment of SCI. In this review, we discuss the role of miRNAs transported by different cell-derived exosomes in SCI that are poised to enhance SCI-specific therapeutic capabilities of exosomes.
Collapse
|
5
|
Zhang Y, Liu J, Xie C, Wu P. Overexpression of miR-133b protects against isoflurane-induced learning and memory impairment. Exp Ther Med 2021; 22:1207. [PMID: 34584552 DOI: 10.3892/etm.2021.10641] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 11/17/2020] [Indexed: 12/21/2022] Open
Abstract
A number of microRNAs (miRs) have been identified as being involved in the regulation of anesthesia-induced cognitive impairment. The aim of the present study was to investigated the role and potential mechanism of miR-133b in isoflurane-induced learning and memory impairment. An animal model of isoflurane exposure was established using neonatal Sprague-Dawley rats. The rats were trained for Morris water maze (MWM) testing to assess their spatial learning and memory ability. Reverse transcription-quantitative polymerase chain reaction was used for the measurement of miR-133b expression in hippocampal tissues and primary hippocampal neuron cultures. Cell viability was assessed using a Cell Counting Kit-8 assay, and flow cytometric analysis was used to determine the rate of apoptosis. The MWM test results indicated that during the training period, the time required to locate the platform was significantly increased for rats exposed to isoflurane, and this increased time was reduced by the overexpression of miR-133b. The results of a probe trial indicated that isoflurane exposure increased escape latency and decreased the time spent in the platform area for isoflurane-treated rats; however, these effects were reversed by the injection of miR-133b agomir. The in vitro experiments demonstrated that the overexpression of miR-133b attenuated the reduction of neuronal cell viability induced by isoflurane, and inhibited the isoflurane-induced apoptosis of hippocampal neurons. In conclusion, the present study revealed that the overexpression of miR-133b attenuated isoflurane-induced learning and memory impairment in rats. Furthermore, miR-133b overexpression promoted the viability of hippocampal neurons and their resistance to apoptosis when exposed to isoflurane.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Anesthesiology, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Jinyong Liu
- Department of Anesthesiology, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Cuili Xie
- Department of Anesthesiology, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Pingping Wu
- Clinical Laboratory, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| |
Collapse
|
6
|
Qin LX, Tan JQ, Zhang HN, Tang JG, Jiang B, Shen XM, Guo JF, Tan LM, Tang B, Wang CY. Preliminary Study of hsa-mir-626 Change in the Cerebrospinal Fluid in Parkinson's Disease. Neurol India 2021; 69:115-118. [PMID: 33642281 DOI: 10.4103/0028-3886.310102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Context A host of microRNAs have been reported to suppress tumor growth, invasion, and metastasis and play roles in neurodegeneration disorders. Moreover, microRNA changes are found in the peripheral blood, cerebrospinal fluid (CSF), and brain tissues of central nervous system diseases, including glioma, Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis, and depression. Compared with other body fluids, CSF can reflect the brain pathological processes more accurately. Aims To understand whether microRNA expression may be misregulated in patients with PD, and further discover potential diagnostic biomarkers and promising therapeutic targets for PD. Materials and Methods Here, through real-time reverse-transcription polymerase chain reaction (RT-PCR), we compared CSF microRNA from 15 PD patients, 11 AD patients, and 16 controls with other neurologic disorders, such as encephalitis and Guillain-Barre syndrome. Results Finally, we identified hsa-miR-626 changes in the CSF of PD patients. The mean expression level of hsa-miR-626 was significantly reduced in the CSF of PD patients compared with AD patients and controls. Conclusions Our approach provides a preliminary research for identifying biomarkers in the CSF that could be used for the detection, diagnosis, and monitoring of PD.
Collapse
Affiliation(s)
- Li-Xia Qin
- Department of Neurology, The Second Xiangya Hospital, Changsha, China
| | - Jie-Qiong Tan
- State Key Laboratory of Medical Genetics, Changsha, China
| | - Hai-Nan Zhang
- Department of Neurology, The Second Xiangya Hospital, Changsha, China
| | - Jian-Guang Tang
- Department of Neurology, The Second Xiangya Hospital, Changsha, China
| | - Bo Jiang
- Department of Neurology, The Second Xiangya Hospital, Changsha, China
| | - Xiang-Min Shen
- Department of Neurology, The Second Xiangya Hospital, Changsha, China
| | - Ji-Feng Guo
- State Key Laboratory of Medical Genetics; Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Li-Ming Tan
- Department of Neurology, The Second Xiangya Hospital, Changsha, China
| | - Beisha Tang
- State Key Laboratory of Medical Genetics; Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Chun-Yu Wang
- Department of Neurology, The Second Xiangya Hospital, Changsha, China
| |
Collapse
|
7
|
Giagnorio E, Malacarne C, Mantegazza R, Bonanno S, Marcuzzo S. MyomiRs and their multifaceted regulatory roles in muscle homeostasis and amyotrophic lateral sclerosis. J Cell Sci 2021; 134:269129. [PMID: 34137441 DOI: 10.1242/jcs.258349] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by loss of both upper and lower motor neurons (MNs). The main clinical features of ALS are motor function impairment, progressive muscle weakness, muscle atrophy and, ultimately, paralysis. Intrinsic skeletal muscle deterioration plays a crucial role in the disease and contributes to ALS progression. Currently, there are no effective treatments for ALS, highlighting the need to obtain a deeper understanding of the molecular events underlying degeneration of both MNs and muscle tissue, with the aim of developing successful therapies. Muscle tissue is enriched in a group of microRNAs called myomiRs, which are effective regulators of muscle homeostasis, plasticity and myogenesis in both physiological and pathological conditions. After providing an overview of ALS pathophysiology, with a focus on the role of skeletal muscle, we review the current literature on myomiR network dysregulation as a contributing factor to myogenic perturbations and muscle atrophy in ALS. We argue that, in view of their critical regulatory function at the interface between MNs and skeletal muscle fiber, myomiRs are worthy of further investigation as potential molecular targets of therapeutic strategies to improve ALS symptoms and counteract disease progression.
Collapse
Affiliation(s)
- Eleonora Giagnorio
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy.,PhD program in Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Claudia Malacarne
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy.,PhD program in Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Renato Mantegazza
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Silvia Bonanno
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Stefania Marcuzzo
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| |
Collapse
|
8
|
Liu WZ, Ma ZJ, Li JR, Kang XW. Mesenchymal stem cell-derived exosomes: therapeutic opportunities and challenges for spinal cord injury. Stem Cell Res Ther 2021; 12:102. [PMID: 33536064 PMCID: PMC7860030 DOI: 10.1186/s13287-021-02153-8] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/07/2021] [Indexed: 12/31/2022] Open
Abstract
Spinal cord injury (SCI) often leads to serious motor and sensory dysfunction of the limbs below the injured segment. SCI not only results in physical and psychological harm to patients but can also cause a huge economic burden on their families and society. As there is no effective treatment method, the prevention, treatment, and rehabilitation of patients with SCI have become urgent problems to be solved. In recent years, mesenchymal stem cells (MSCs) have attracted more attention in the treatment of SCI. Although MSC therapy can reduce injured volume and promote axonal regeneration, its application is limited by tumorigenicity, a low survival rate, and immune rejection. Accumulating literature shows that exosomes have great potential in the treatment of SCI. In this review, we summarize the existing MSC-derived exosome studies on SCI and discuss the advantages and challenges of treating SCI based on exosomes derived from MSCs.
Collapse
Affiliation(s)
- Wen-Zhao Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, China
- Department of Orthopedics, Lanzhou University Second Hospital, No.82 Cuiyingmen Street, Lanzhou, 730030, Gansu, China
| | - Zhan-Jun Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, China
- Department of Orthopedics, Lanzhou University Second Hospital, No.82 Cuiyingmen Street, Lanzhou, 730030, Gansu, China
| | - Jie-Ru Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Xue-Wen Kang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, China.
- Department of Orthopedics, Lanzhou University Second Hospital, No.82 Cuiyingmen Street, Lanzhou, 730030, Gansu, China.
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
9
|
Taetzsch T, Shapiro D, Eldosougi R, Myers T, Settlage RE, Valdez G. The microRNA miR-133b functions to slow Duchenne muscular dystrophy pathogenesis. J Physiol 2021; 599:171-192. [PMID: 32991751 PMCID: PMC8418193 DOI: 10.1113/jp280405] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/14/2020] [Indexed: 01/22/2023] Open
Abstract
KEY POINTS Impairment of muscle biogenesis contributes to the progression of Duchenne muscular dystrophy (DMD). As a muscle enriched microRNA that has been implicated in muscle biogenesis, the role of miR-133b in DMD remains unknown. To assess miR-133b function in DMD-affected skeletal muscles, we genetically ablated miR-133b in the mdx mouse model of DMD. We show that deletion of miR-133b exacerbates the dystrophic phenotype of DMD-afflicted skeletal muscle by dysregulating muscle stem cells involved in muscle biogenesis, in addition to affecting signalling pathways related to inflammation and fibrosis. Our results provide evidence that miR-133b may underlie DMD pathology by affecting the proliferation and differentiation of muscle stem cells. ABSTRACT Duchenne muscular dystrophy (DMD) is characterized by progressive skeletal muscle degeneration. No treatments are currently available to prevent the disease. While the muscle enriched microRNA miR-133b has been implicated in muscle biogenesis, its role in DMD remains unknown. To assess miR-133b function in DMD-affected skeletal muscles, we genetically ablated miR-133b in the mdx mouse model of DMD. In the absence of miR-133b, the tibialis anterior muscle of P30 mdx mice is smaller in size and exhibits a thickened interstitial space containing more mononucleated cells. Additional analysis revealed that miR-133b deletion influences muscle fibre regeneration, satellite cell proliferation and differentiation, and induces widespread transcriptomic changes in mdx muscle. These include known miR-133b targets as well as genes involved in cell proliferation and fibrosis. Altogether, our data demonstrate that skeletal muscles utilize miR-133b to mitigate the deleterious effects of DMD.
Collapse
Affiliation(s)
- Thomas Taetzsch
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Dillon Shapiro
- Molecular Biology, Cell Biology, & Biochemistry Graduate Program, Brown University, Providence, RI, USA
| | - Randa Eldosougi
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Tracey Myers
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | | | - Gregorio Valdez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, United States
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, United States
| |
Collapse
|
10
|
Ardashirova NS, Fedotova EY, Illarioshkin SN. The Role of MicroRNA in the Pathogenesis and Diagnostics of Parkinson’s Disease. NEUROCHEM J+ 2020. [DOI: 10.1134/s1819712420020026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
11
|
Mak HK, Yung JSY, Weinreb RN, Ng SH, Cao X, Ho TYC, Ng TK, Chu WK, Yung WH, Choy KW, Wang CC, Lee TL, Leung CKS. MicroRNA-19a-PTEN Axis Is Involved in the Developmental Decline of Axon Regenerative Capacity in Retinal Ganglion Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 21:251-263. [PMID: 32599451 PMCID: PMC7327411 DOI: 10.1016/j.omtn.2020.05.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/15/2020] [Accepted: 05/28/2020] [Indexed: 11/23/2022]
Abstract
Irreversible blindness from glaucoma and optic neuropathies is attributed to retinal ganglion cells (RGCs) losing the ability to regenerate axons. While several transcription factors and proteins have demonstrated enhancement of axon regeneration after optic nerve injury, mechanisms contributing to the age-related decline in axon regenerative capacity remain elusive. In this study, we show that microRNAs are differentially expressed during RGC development and identify microRNA-19a (miR-19a) as a heterochronic marker; developmental decline of miR-19a relieves suppression of phosphatase and tensin homolog (PTEN), a key regulator of axon regeneration, and serves as a temporal indicator of decreasing axon regenerative capacity. Intravitreal injection of miR-19a promotes axon regeneration after optic nerve crush in adult mice, and it increases axon extension in RGCs isolated from aged human donors. This study uncovers a previously unrecognized involvement of the miR-19a-PTEN axis in RGC axon regeneration, and it demonstrates therapeutic potential of microRNA-mediated restoration of axon regenerative capacity in optic neuropathies.
Collapse
Affiliation(s)
- Heather K Mak
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Jasmine S Y Yung
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Robert N Weinreb
- Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, La Jolla, CA, USA; Department of Ophthalmology, University of California, San Diego, La Jolla, CA, USA
| | - Shuk Han Ng
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Xu Cao
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Tracy Y C Ho
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Tsz Kin Ng
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Wai Kit Chu
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Wing Ho Yung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PRC; Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Kwong Wai Choy
- Department of Obstetrics and Gynecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Chi Chiu Wang
- Department of Obstetrics and Gynecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, PRC
| | - Tin Lap Lee
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PRC
| | | |
Collapse
|
12
|
Preliminary study of hsa-miR-626 change in the cerebrospinal fluid of Parkinson’s disease patients. J Clin Neurosci 2019; 70:198-201. [DOI: 10.1016/j.jocn.2019.08.082] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 07/19/2019] [Accepted: 08/16/2019] [Indexed: 12/29/2022]
|
13
|
MicroRNAs: Game Changers in the Regulation of α-Synuclein in Parkinson's Disease. PARKINSONS DISEASE 2019; 2019:1743183. [PMID: 31191899 PMCID: PMC6525811 DOI: 10.1155/2019/1743183] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 03/24/2019] [Accepted: 04/10/2019] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder. Its neuropathological hallmarks include neuronal loss in the substantia nigra pars compacta (SNpc) and the presence of Lewy bodies containing aggregates of α-synuclein (α-syn). An imbalance between the rates of α-syn synthesis, aggregation, and clearance can result in abnormal α-syn levels and contribute to the pathogenesis of PD. MicroRNAs (miRNAs) are endogenous single-stranded noncoding RNAs (∼22 nucleotides) that have recently emerged as key posttranscriptional regulators of gene expression. In this review, we summarize the functions of miRNAs that directly target α-syn. We also review miRNAs that indirectly impact α-syn levels or toxicity through different pathways, including those involved in the clearance of α-syn and neuroinflammation.
Collapse
|
14
|
Sarkar SN, Russell AE, Engler-Chiurazzi EB, Porter KN, Simpkins JW. MicroRNAs and the Genetic Nexus of Brain Aging, Neuroinflammation, Neurodegeneration, and Brain Trauma. Aging Dis 2019; 10:329-352. [PMID: 31011481 PMCID: PMC6457055 DOI: 10.14336/ad.2018.0409] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 04/09/2018] [Indexed: 12/12/2022] Open
Abstract
Aging is a complex and integrated gradual deterioration of cellular activities in specific organs of the body, which is associated with increased mortality. This deterioration is the primary risk factor for major human pathologies, including cancer, diabetes, cardiovascular disorders, neurovascular disorders, and neurodegenerative diseases. There are nine tentative hallmarks of aging. In addition, several of these hallmarks are increasingly being associated with acute brain injury conditions. In this review, we consider the genes and their functional pathways involved in brain aging as a means of developing new strategies for therapies targeted to the neuropathological processes themselves, but also as targets for many age-related brain diseases. A single microRNA (miR), which is a short, non-coding RNA species, has the potential for targeting many genes simultaneously and, like practically all other cellular processes, genes associated with many features of brain aging and injury are regulated by miRs. We highlight how certain miRs can mediate deregulation of genes involved in neuroinflammation, acute neuronal injury and chronic neurodegenerative diseases. Finally, we review the recent progress in the development of effective strategies to block specific miR functions and discuss future approaches with the prediction that anti-miR drugs may soon be used in the clinic.
Collapse
Affiliation(s)
- Saumyendra N Sarkar
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Ashley E Russell
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Elizabeth B Engler-Chiurazzi
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Keyana N Porter
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - James W Simpkins
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
15
|
Vidal PM, Pacheco R. Targeting the Dopaminergic System in Autoimmunity. J Neuroimmune Pharmacol 2019; 15:57-73. [PMID: 30661214 DOI: 10.1007/s11481-019-09834-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023]
Abstract
Dopamine has emerged as a fundamental regulator of inflammation. In this regard, it has been shown that dopaminergic signalling pathways are key players promoting homeostasis between the central nervous system and the immune system. Dysregulation in the dopaminergic system affects both innate and adaptive immunity, contributing to the development of numerous autoimmune and inflammatory pathologies. This makes dopamine receptors interesting therapeutic targets for either the development of new treatments or repurposing of already available pharmacological drugs. Dopamine receptors are broadly expressed on different immune cells with multifunctional effects depending on the dopamine concentration available and the pattern of expression of five dopamine receptors displaying different affinities for dopamine. Thus, impaired dopaminergic signalling through different dopamine receptors may result in altered behaviour of immunity, contributing to the development and progression of autoimmune pathologies. In this review we discuss the current evidence involving the dopaminergic system in inflammatory bowel disease, multiple sclerosis and Parkinson's disease. In addition, we summarise and analyse the therapeutic approaches designed to attenuate disease development and progression by targeting the dopaminergic system. Graphical Abstract Targetting the dopaminergic system in autoimmunity. Effector T-cells (Teff) orchestrate inflamamtion involved in autoimmunity, whilst regulatory T-cells (Tregs) suppress Teff activity promoting tolerance to self-constituents. Dopamine has emerged as a key regulator of Teff and Tregs function, thereby dopamine receptors have becoming important therapeutic targets in autoimmune disorders, especially in those affecting the brain and the gut, where dopamine levels strongly change with inflammation.
Collapse
Affiliation(s)
- Pia M Vidal
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Av. Zañartu 1482, Ñuñoa, 7780272, Santiago, Chile
| | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Av. Zañartu 1482, Ñuñoa, 7780272, Santiago, Chile. .,Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, 8370146, Santiago, Chile.
| |
Collapse
|
16
|
Li D, Zhang P, Yao X, Li H, Shen H, Li X, Wu J, Lu X. Exosomes Derived From miR-133b-Modified Mesenchymal Stem Cells Promote Recovery After Spinal Cord Injury. Front Neurosci 2018; 12:845. [PMID: 30524227 PMCID: PMC6262643 DOI: 10.3389/fnins.2018.00845] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/29/2018] [Indexed: 12/30/2022] Open
Abstract
Dysregulation of microRNAs (miRNAs) has been found in injured spinal cords after spinal cord injury (SCI). Previous studies have shown that miR-133b plays an important role in the differentiation of neurons and the outgrowth of neurites. Recently, exosomes have been used as novel biological vehicles to transfer miRNAs locally or systemically, but little is known about the effect of the delivery of exosome-mediated miRNAs on the treatment of SCI. In the present study, we observed that mesenchymal stem cells, the most common cell types known to produce exosomes, could package miR-133b into secreted exosomes. After SCI, tail vein injection of miR-133b exosomes into rats significantly improved the recovery of hindlimb function when compared to control groups. Additionally, treatment with miR-133b exosomes reduced the volume of the lesion, preserved neuronal cells, and promoted the regeneration of axons after SCI. We next observed that the expression of RhoA, a direct target of miR-133b, was decreased in the miR-133b exosome group. Moreover, we showed that miR-133b exosomes activated ERK1/2, STAT3, and CREB, which are signaling pathway proteins involved in the survival of neurons and the regeneration of axons. In summary, these findings demonstrated that systemically injecting miR-133b exosomes preserved neurons, promoted the regeneration of axons, and improved the recovery of hindlimb locomotor function following SCI, suggesting that the transfer of exosome-mediated miRNAs represents a novel therapeutic approach for the treatment of SCI.
Collapse
Affiliation(s)
- Dong Li
- Department of Neurosurgery, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, China
| | - Peng Zhang
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiyang Yao
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haiying Li
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haitao Shen
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiang Li
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiang Wu
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaocheng Lu
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
17
|
Leggio L, Vivarelli S, L'Episcopo F, Tirolo C, Caniglia S, Testa N, Marchetti B, Iraci N. microRNAs in Parkinson's Disease: From Pathogenesis to Novel Diagnostic and Therapeutic Approaches. Int J Mol Sci 2017; 18:ijms18122698. [PMID: 29236052 PMCID: PMC5751299 DOI: 10.3390/ijms18122698] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/07/2017] [Accepted: 12/09/2017] [Indexed: 01/09/2023] Open
Abstract
Parkinson’s disease (PD) is the most prevalent central nervous system (CNS) movement disorder and the second most common neurodegenerative disease overall. PD is characterized by the progressive loss of dopaminergic (DAergic) neurons in the substantia nigra pars compacta (SNpc) within the midbrain, accumulation of alpha-synuclein (α-SYN) in Lewy bodies and neurites and excessive neuroinflammation. The neurodegenerative processes typically begin decades before the appearance of clinical symptoms. Therefore, the diagnosis is achievable only when the majority of the relevant DAergic neurons have already died and for that reason available treatments are only palliative at best. The causes and mechanism(s) of this devastating disease are ill-defined but complex interactions between genetic susceptibility and environmental factors are considered major contributors to the etiology of PD. In addition to the role of classical gene mutations in PD, the importance of regulatory elements modulating gene expression has been increasingly recognized. One example is the critical role played by microRNAs (miRNAs) in the development and homeostasis of distinct populations of neurons within the CNS and, in particular, in the context of PD. Recent reports demonstrate how distinct miRNAs are involved in the regulation of PD genes, whereas profiling approaches are unveiling variations in the abundance of certain miRNAs possibly relevant either to the onset or to the progression of the disease. In this review, we provide an overview of the miRNAs recently found to be implicated in PD etiology, with particular focus on their potential relevance as PD biomarkers, as well as their possible use in PD targeted therapy.
Collapse
Affiliation(s)
- Loredana Leggio
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via S. Sofia 97, 95125 Catania, Italy.
| | - Silvia Vivarelli
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via S. Sofia 97, 95125 Catania, Italy.
| | - Francesca L'Episcopo
- Neuropharmacology Section, OASI Institute for Research and Care on Mental Retardation and Brain Aging (IRCCS), 94018 Troina, Italy.
| | - Cataldo Tirolo
- Neuropharmacology Section, OASI Institute for Research and Care on Mental Retardation and Brain Aging (IRCCS), 94018 Troina, Italy.
| | - Salvo Caniglia
- Neuropharmacology Section, OASI Institute for Research and Care on Mental Retardation and Brain Aging (IRCCS), 94018 Troina, Italy.
| | - Nunzio Testa
- Neuropharmacology Section, OASI Institute for Research and Care on Mental Retardation and Brain Aging (IRCCS), 94018 Troina, Italy.
| | - Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via S. Sofia 97, 95125 Catania, Italy.
- Neuropharmacology Section, OASI Institute for Research and Care on Mental Retardation and Brain Aging (IRCCS), 94018 Troina, Italy.
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via S. Sofia 97, 95125 Catania, Italy.
| |
Collapse
|
18
|
Huang R, Chen M, Yang L, Wagle M, Guo S, Hu B. MicroRNA-133b Negatively Regulates Zebrafish Single Mauthner-Cell Axon Regeneration through Targeting tppp3 in Vivo. Front Mol Neurosci 2017; 10:375. [PMID: 29209165 PMCID: PMC5702462 DOI: 10.3389/fnmol.2017.00375] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 10/27/2017] [Indexed: 12/30/2022] Open
Abstract
Axon regeneration, fundamental to nerve repair, and functional recovery, relies on rapid changes in gene expression attributable to microRNA (miRNA) regulation. MiR-133b has been proved to play an important role in different organ regeneration in zebrafish, but its role in regulating axon regeneration in vivo is still controversial. Here, combining single-cell electroporation with a vector-based miRNA-expression system, we have modulated the expression of miR-133b in Mauthner-cells (M-cells) at the single-cell level in zebrafish. Through in vivo imaging, we show that overexpression of miR-133b inhibits axon regeneration, whereas down-regulation of miR-133b, promotes axon outgrowth. We further show that miR-133b regulates axon regeneration by directly targeting a novel regeneration-associated gene, tppp3, which belongs to Tubulin polymerization-promoting protein family. Gain or loss-of-function of tppp3 experiments indicated that tppp3 was a novel gene that could promote axon regeneration. In addition, we observed a reduction of mitochondrial motility, which have been identified to have a positive correlation with axon regeneration, in miR-133b overexpressed M-cells. Taken together, our work provides a novel way to study the role of miRNAs in individual cell and establishes a critical cell autonomous role of miR-133b in zebrafish M-cell axon regeneration. We propose that up-regulation of the newly founded regeneration-associated gene tppp3 may enhance axonal regeneration.
Collapse
Affiliation(s)
- Rongchen Huang
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Min Chen
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Leiqing Yang
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Mahendra Wagle
- Programs in Human Genetics and Biological Sciences, Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Su Guo
- Programs in Human Genetics and Biological Sciences, Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Bing Hu
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China
| |
Collapse
|
19
|
Astroglial MicroRNA-219-5p in the Ventral Tegmental Area Regulates Nociception in Rats. Anesthesiology 2017; 127:548-564. [PMID: 28582325 DOI: 10.1097/aln.0000000000001720] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND The authors previously reported that noncoding microRNA miR-219-5p is down-regulated in the spinal cord in a nociceptive state. The ventral tegmental area also plays critical roles in modulating nociception, although the underlying mechanism remains unknown. The authors hypothesized that miR-219-5p in the ventral tegmental area also may modulate nociception. METHODS The authors studied the bidirectional regulatory role of ventral tegmental area miR-219-5p in a rat complete Freund's adjuvant model of inflammatory nociception by measuring paw withdrawal latencies. Using molecular biology technologies, the authors measured the effects of astroglial coiled-coil and C2 domain containing 1A/nuclear factor κB cascade and dopamine neuron activity on the down-regulation of ventral tegmental area miR-219-5p-induced nociceptive responses. RESULTS MiR-219-5p expression in the ventral tegmental area was reduced in rats with thermal hyperalgesia. Viral overexpression of ventral tegmental area miR-219-5p attenuated complete Freund's adjuvant-induced nociception from 7 days after complete Freund's adjuvant injection (paw withdrawal latencies: 6.09 ± 0.83 s vs. 3.96 ± 0.76 s; n = 6/group). Down-regulation of ventral tegmental area miR-219-5p in naïve rats was sufficient to induce thermal hyperalgesia from 7 days after lentivirus injection (paw withdrawal latencies: 7.09 ± 1.54 s vs. 11.75 ± 2.15 s; n = 8/group), which was accompanied by increased glial fibrillary acidic protein (fold change: 2.81 ± 0.38; n = 3/group) and reversed by intraventral tegmental area injection of the astroglial inhibitor fluorocitrate. The nociceptive responses induced by astroglial miR-219-5p down-regulation were inhibited by interfering with astroglial coiled-coil and C2 domain containing 1A/nuclear factor-κB signaling. Finally, pharmacologic inhibition of ventral tegmental area dopamine neurons alleviated this hyperalgesia. CONCLUSIONS Down-regulation of astroglial miR-219-5p in ventral tegmental area induced nociceptive responses are mediated by astroglial coiled-coil and C2 domain containing 1A/nuclear factor-κB signaling and elevated dopamine neuron activity.
Collapse
|
20
|
van der Stijl R, Withoff S, Verbeek DS. Spinocerebellar ataxia: miRNAs expose biological pathways underlying pervasive Purkinje cell degeneration. Neurobiol Dis 2017; 108:148-158. [PMID: 28823930 DOI: 10.1016/j.nbd.2017.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/21/2017] [Accepted: 08/16/2017] [Indexed: 01/09/2023] Open
Abstract
Recent work has demonstrated the importance of miRNAs in the pathogenesis of various brain disorders including the neurodegenerative disorder spinocerebellar ataxia (SCA). This review focuses on the role of miRNAs in the shared pathogenesis of the different SCA types. We examine the novel findings of a recent cell-type-specific RNA-sequencing study in mouse brain and discuss how the identification of Purkinje-cell-enriched miRNAs highlights biological pathways that expose the mechanisms behind pervasive Purkinje cell degeneration in SCA. These key pathways are likely to contain targets for therapeutic development and represent potential candidate genes for genetically unsolved SCAs.
Collapse
Affiliation(s)
- Rogier van der Stijl
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Sebo Withoff
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Dineke S Verbeek
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.
| |
Collapse
|
21
|
Rajman M, Schratt G. MicroRNAs in neural development: from master regulators to fine-tuners. Development 2017; 144:2310-2322. [DOI: 10.1242/dev.144337] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The proper formation and function of neuronal networks is required for cognition and behavior. Indeed, pathophysiological states that disrupt neuronal networks can lead to neurodevelopmental disorders such as autism, schizophrenia or intellectual disability. It is well-established that transcriptional programs play major roles in neural circuit development. However, in recent years, post-transcriptional control of gene expression has emerged as an additional, and probably equally important, regulatory layer. In particular, it has been shown that microRNAs (miRNAs), an abundant class of small regulatory RNAs, can regulate neuronal circuit development, maturation and function by controlling, for example, local mRNA translation. It is also becoming clear that miRNAs are frequently dysregulated in neurodevelopmental disorders, suggesting a role for miRNAs in the etiology and/or maintenance of neurological disease states. Here, we provide an overview of the most prominent regulatory miRNAs that control neural development, highlighting how they act as ‘master regulators’ or ‘fine-tuners’ of gene expression, depending on context, to influence processes such as cell fate determination, cell migration, neuronal polarization and synapse formation.
Collapse
Affiliation(s)
- Marek Rajman
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-Universität Marburg, Marburg 35043, Germany
| | - Gerhard Schratt
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-Universität Marburg, Marburg 35043, Germany
| |
Collapse
|
22
|
Zhang X, Yang R, Hu BL, Lu P, Zhou LL, He ZY, Wu HM, Zhu JH. Reduced Circulating Levels of miR-433 and miR-133b Are Potential Biomarkers for Parkinson's Disease. Front Cell Neurosci 2017; 11:170. [PMID: 28690499 PMCID: PMC5481393 DOI: 10.3389/fncel.2017.00170] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/06/2017] [Indexed: 11/15/2022] Open
Abstract
Aberrant expression of microRNA (miRNA) in tissues may lead to altered level in circulation. Considerable evidence has suggested that miRNA deregulation is involved in the pathogenesis of Parkinson’s disease (PD). In this study, we screened a set of PD-associated miRNAs and aimed to identify differentially expressed miRNAs in plasma of PD patients and to evaluate their potentiality to serve as PD biomarkers. A total of 95 subjects consisting of 46 sporadic PD cases and 49 controls were recruited. Plasma levels of six miRNAs including miR-433, miR-133b, miR-34b, miR-34c, miR-153, and miR-7 were evaluated using reverse transcribed quantitative PCR, among which we found that miR-34c and miR-7 were below detection limit under our condition. The results showed that levels of circulating miR-433 (P = 0.003) and miR-133b (P = 0.006), but not miR-34b and miR-153, were reduced in PD patients. miR-433 and miR-133b were strongly correlated in both control and PD groups (rs = 0.87 and 0.85, respectively). The correlation between miR-34b and miR-153 expressions was significantly reduced (P < 0.05) in the PD group. Although miR-433 and miR-133b were likely to be functionally complimentary as suggested by Pathway and Gene Ontology analyses, these two miRNAs per se might not be sufficient to predict PD. No correlation was observed between the four miRNAs and age or severity of disease. Collectively, our results demonstrate that circulating miR-433 and miR-133b are significantly altered in PD and may serve as PD biomarkers.
Collapse
Affiliation(s)
- Xiong Zhang
- Department of Geriatrics and Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, WenzhouChina.,Department of Preventive Medicine, Wenzhou Medical University, WenzhouChina
| | - Rui Yang
- Department of Preventive Medicine, Wenzhou Medical University, WenzhouChina
| | - Bei-Lei Hu
- Department of Geriatrics and Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, WenzhouChina
| | - Pengcheng Lu
- Department of Biostatistics Graduate Program, University of Kansas Medical Center, Kansas CityKS, United States
| | - Li-Li Zhou
- Department of Preventive Medicine, Wenzhou Medical University, WenzhouChina
| | - Zhi-Yong He
- Department of Geriatrics and Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, WenzhouChina
| | - Hong-Mei Wu
- Department of Preventive Medicine, Wenzhou Medical University, WenzhouChina
| | - Jian-Hong Zhu
- Department of Geriatrics and Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, WenzhouChina.,Department of Preventive Medicine, Wenzhou Medical University, WenzhouChina.,Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, WenzhouChina
| |
Collapse
|
23
|
Singh A, Sen D. MicroRNAs in Parkinson's disease. Exp Brain Res 2017; 235:2359-2374. [PMID: 28526930 DOI: 10.1007/s00221-017-4989-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 05/16/2017] [Indexed: 01/11/2023]
Abstract
Parkinson's disease is the second most common neurodegenerative disease commonly affecting the older population. Loss of dopaminergic neurons in the substantia nigra of brain leads to impairment of motor activities as well as cognitive defects. There are many underlying causes to this disease, both genetic and epigenetic, which are yet to be fully explored. Non-coding RNAs are significant part of our genome and are involved in various cellular processes. MicroRNAs, which are small non-coding RNAs having 20-22 nucleotides, are involved in many underlying mechanisms of pathogenesis of several neurodegenerative diseases including Parkinson's. This review focuses on the role played by microRNAs in regulating various genes responsible for the onset and pathogenesis of Parkinson's disease and various literature evidences pointing at the usefulness of targeting specific microRNAs as a potential alternate therapeutic strategy for successful impairment of the disease progression. This review also discusses about various biofluid-based microRNA markers which may be potentially utilized for diagnostic purposes.
Collapse
Affiliation(s)
- Abhishek Singh
- School of Bio Sciences and Technology, VIT University, Vellore, India
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), VIT University, Vellore, Tamil Nadu, 632014, India
| | - Dwaipayan Sen
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), VIT University, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
24
|
Lenihan JA, Saha O, Heimer-McGinn V, Cryan JF, Feng G, Young PW. Decreased Anxiety-Related Behaviour but Apparently Unperturbed NUMB Function in Ligand of NUMB Protein-X (LNX) 1/2 Double Knockout Mice. Mol Neurobiol 2016; 54:8090-8109. [DOI: 10.1007/s12035-016-0261-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 10/25/2016] [Indexed: 10/20/2022]
|
25
|
Ding HL, Hooper JE, Batzel P, Eames BF, Postlethwait JH, Artinger KB, Clouthier DE. MicroRNA Profiling during Craniofacial Development: Potential Roles for Mir23b and Mir133b. Front Physiol 2016; 7:281. [PMID: 27471470 PMCID: PMC4943961 DOI: 10.3389/fphys.2016.00281] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 06/21/2016] [Indexed: 01/01/2023] Open
Abstract
Defects in mid-facial development, including cleft lip/palate, account for a large number of human birth defects annually. In many cases, aberrant gene expression results in either a reduction in the number of neural crest cells (NCCs) that reach the frontonasal region and form much of the facial skeleton or subsequent failure of NCC patterning and differentiation into bone and cartilage. While loss of gene expression is often associated with developmental defects, aberrant upregulation of expression can also be detrimental. microRNAs (miRNAs) are a class of non-coding RNAs that normally repress gene expression by binding to recognition sequences located in the 3′ UTR of target mRNAs. miRNAs play important roles in many developmental systems, including midfacial development. Here, we take advantage of high throughput RNA sequencing (RNA-seq) from different tissues of the developing mouse midface to interrogate the miRs that are expressed in the midface and select a subset for further expression analysis. Among those examined, we focused on four that showed the highest expression level in in situ hybridization analysis. Mir23b and Mir24.1 are specifically expressed in the developing mouse frontonasal region, in addition to areas in the perichondrium, tongue musculature and cranial ganglia. Mir23b is also expressed in the palatal shelves and in anterior epithelium of the palate. In contrast, Mir133b and Mir128.2 are mainly expressed in head and trunk musculature. Expression analysis of mir23b and mir133b in zebrafish suggests that mir23b is expressed in the pharyngeal arch, otic vesicle, and trunk muscle while mir133b is similarly expressed in head and trunk muscle. Functional analysis by overexpression of mir23b in zebrafish leads to broadening of the ethmoid plate and aberrant cartilage structures in the viscerocranium, while overexpression of mir133b causes a reduction in ethmoid plate size and a significant midfacial cleft. These data illustrate that miRs are expressed in the developing midface and that Mir23b and Mir133b may have roles in this developmental process.
Collapse
Affiliation(s)
- Hai-Lei Ding
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - Joan E Hooper
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - Peter Batzel
- Department of Neuroscience, University of Oregon Eugene, OR, USA
| | - B Frank Eames
- Department of Neuroscience, University of OregonEugene, OR, USA; Department of Anatomy and Cell Biology, University of SaskatchewanSaskatoon, SK, Canada
| | | | - Kristin B Artinger
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - David E Clouthier
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| |
Collapse
|
26
|
Zhang Q, Gao X, Li C, Feliciano C, Wang D, Zhou D, Mei Y, Monteiro P, Anand M, Itohara S, Dong X, Fu Z, Feng G. Impaired Dendritic Development and Memory in Sorbs2 Knock-Out Mice. J Neurosci 2016; 36:2247-60. [PMID: 26888934 PMCID: PMC4756157 DOI: 10.1523/jneurosci.2528-15.2016] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 12/21/2015] [Accepted: 01/13/2016] [Indexed: 12/27/2022] Open
Abstract
Intellectual disability is a common neurodevelopmental disorder characterized by impaired intellectual and adaptive functioning. Both environmental insults and genetic defects contribute to the etiology of intellectual disability. Copy number variations of SORBS2 have been linked to intellectual disability. However, the neurobiological function of SORBS2 in the brain is unknown. The SORBS2 gene encodes ArgBP2 (Arg/c-Abl kinase binding protein 2) protein in non-neuronal tissues and is alternatively spliced in the brain to encode nArgBP2 protein. We found nArgBP2 colocalized with F-actin at dendritic spines and growth cones in cultured hippocampal neurons. In the mouse brain, nArgBP2 was highly expressed in the cortex, amygdala, and hippocampus, and enriched in the outer one-third of the molecular layer in dentate gyrus. Genetic deletion of Sorbs2 in mice led to reduced dendritic complexity and decreased frequency of AMPAR-miniature spontaneous EPSCs in dentate gyrus granule cells. Behavioral characterization revealed that Sorbs2 deletion led to a reduced acoustic startle response, and defective long-term object recognition memory and contextual fear memory. Together, our findings demonstrate, for the first time, an important role for nArgBP2 in neuronal dendritic development and excitatory synaptic transmission, which may thus inform exploration of neurobiological basis of SORBS2 deficiency in intellectual disability. SIGNIFICANCE STATEMENT Copy number variations of the SORBS2 gene are linked to intellectual disability, but the neurobiological mechanisms are unknown. We found that nArgBP2, the only neuronal isoform encoded by SORBS2, colocalizes with F-actin at neuronal dendritic growth cones and spines. nArgBP2 is highly expressed in the cortex, amygdala, and dentate gyrus in the mouse brain. Genetic deletion of Sorbs2 in mice leads to impaired dendritic complexity and reduced excitatory synaptic transmission in dentate gyrus granule cells, accompanied by behavioral deficits in acoustic startle response and long-term memory. This is the first study of Sorbs2 function in the brain, and our findings may facilitate the study of neurobiological mechanisms underlying SORBS2 deficiency in the development of intellectual disability.
Collapse
Affiliation(s)
- Qiangge Zhang
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Xian Gao
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Key Laboratory of Brain Functional Genomics (Ministry of Education and Science and Technology Commission of Shanghai Municipality), Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Chenchen Li
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Catia Feliciano
- Champalimaud Neuroscience Programme, Champalimaud Center for the Unknown, Lisbon 1400-038, Portugal
| | - Dongqing Wang
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Dingxi Zhou
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, School of Life Sciences, Peking University, Beijing 100871, China, and
| | - Yuan Mei
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Patricia Monteiro
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Michelle Anand
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Shigeyoshi Itohara
- Laboratory of Behavioral Genetics, RIKEN Brain Science Institute, Wako 351-0198, Japan
| | - Xiaowei Dong
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Science and Technology Commission of Shanghai Municipality), Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
| | - Zhanyan Fu
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Key Laboratory of Brain Functional Genomics (Ministry of Education and Science and Technology Commission of Shanghai Municipality), Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142,
| |
Collapse
|
27
|
Saghazadeh A, Rezaei N. MicroRNA machinery in Parkinson's disease: a platform for neurodegenerative diseases. Expert Rev Neurother 2015; 22:427-453. [PMID: 26574782 DOI: 10.1586/14737175.2015.1114886] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
MicroRNAs (miRNAs) are noncoding RNAs that recognize their protein-coding target genes and whereby subjugate them after transcription. Despite the infancy of this field of science, the role of miRNAs in neurodegeneration is well-acknowledged. This review was conducted to indicate that Parkinson's disease (PD) is not excluded from this rule. To this end, we evaluated the existing literature and arranged PD-associated miRNAs according to their mechanism of action, particularly apoptosis, autophagy, inflammation, mitochondrial dysfunction and oxidative stress. According to this arrangement, a majority of PD-associated miRNAs were indicated to influence autophagic/apoptotic pathways. We also categorized PD-associated miRNAs according to that they could exert detrimental or beneficial or both into three sets, activator, inhibitor, and double-edged, correspondingly. Considering this criterion, a majority of PD-associated miRNAs were included in the activator category. In addition, evidences from genetic association studies investigating genetic variants of or related to miRNAs in PD patients are presented. Finally, possible applications of the miRNA machinery in PD, including mechanistic networks, diagnostic, prognostic and therapeutic potentials, are discussed. But there may be additional miRNAs involved in the pathogenesis of PD which have hitherto remained unknown and thus further studies are needed to explore the issue and to extend this platform.
Collapse
Affiliation(s)
- Amene Saghazadeh
- a Molecular Immunology Research Center and Department of Immunology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Nima Rezaei
- a Molecular Immunology Research Center and Department of Immunology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,b Research Center for Immunodeficiencies, Children's Medical Center , Tehran University of Medical Sciences , Tehran , Iran.,c Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| |
Collapse
|
28
|
Zhang Y, Wang Y, Wang L, Bai M, Zhang X, Zhu X. Dopamine Receptor D2 and Associated microRNAs Are Involved in Stress Susceptibility and Resistance to Escitalopram Treatment. Int J Neuropsychopharmacol 2015; 18:pyv025. [PMID: 25740916 PMCID: PMC4571637 DOI: 10.1093/ijnp/pyv025] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Early life stress has been demonstrated to increase the risk of developing depression in adulthood. However, the roles and associated molecular mechanisms of stresses in the onset and relapse of depression have yet to be fully elucidated. METHODS Depression-like behaviors were induced in rats by maternal deprivation and chronic unpredictable stress. Depression- and anxiety-like behaviors of rats, dopamine receptor D2 level, and microRNAs expression in rats' brain tissues were measured. RESULTS Chronic unpredictable stress alone induced depression-like behaviors in rats, but maternal deprivation enhanced the effect of chronic unpredictable stress. Escitalopram significantly decreased depression-like behaviors in chronic unpredictable stress rats but was less effective in maternal deprivation with chronic unpredictable stress rats. Maternal deprivation increased dopamine receptor D2 messenger RNA expression and decreased microRNA-9 expression in the striatum. Chronic unpredictable stress increased dopamine receptor D2 mRNA and protein levels and decreased microRNA-9 expression in the nucleus accumbens. Furthermore, maternal deprivation enhanced the effect of chronic unpredictable stress on dopamine receptor D2 gene and microRNA-9 expression. Chronic unpredictable stress increased the expression of microRNA-326 in the nucleus accumbens but decreased it in the striatum, whereas maternal deprivation elevated microRNA-326 expression in the striatum. Escitalopram normalized microRNA-326 expression but had no effect on the expression of microRNA-9, dopamine receptor D2 mRNA, and dopamine receptor D2 protein in both the nucleus accumbens and striatum. The in vitro study showed that only microRNA-9 directly targeted the 3' untranslated region of dopamine receptor D2 mRNA and inhibited dopamine receptor D2 protein expression. CONCLUSION Early life stress enhanced the susceptibility to late life stress and resistance to escitalopram treatment through decreasing microRNA-9 expression and subsequently upregulating dopamine receptor D2 expression in the nucleus accumbens. microRNA-326 may be a novel target of escitalopram.
Collapse
Affiliation(s)
- Yi Zhang
- Medical Psychological Institute, Second Xiangya Hospital, Central South University,Changsha, PR China (Drs Y. Zhang, Y. Wang, L. Wang, Bai, and Zhu); Department of Radiation Oncology, University of Maryland, Baltimore, MD (Drs Y. Zhang and X. Zhang); National Technology Institute of Psychiatry, Central South University, Changsha, PR China (Drs Y. Wang and L. Wang); Key Laboratory of Psychiatry and Mental Health of Hunan Province, Central South University, Changsha, PR China (Drs Bai and Zhu)
| | - Yuting Wang
- Medical Psychological Institute, Second Xiangya Hospital, Central South University,Changsha, PR China (Drs Y. Zhang, Y. Wang, L. Wang, Bai, and Zhu); Department of Radiation Oncology, University of Maryland, Baltimore, MD (Drs Y. Zhang and X. Zhang); National Technology Institute of Psychiatry, Central South University, Changsha, PR China (Drs Y. Wang and L. Wang); Key Laboratory of Psychiatry and Mental Health of Hunan Province, Central South University, Changsha, PR China (Drs Bai and Zhu)
| | - Lei Wang
- Medical Psychological Institute, Second Xiangya Hospital, Central South University,Changsha, PR China (Drs Y. Zhang, Y. Wang, L. Wang, Bai, and Zhu); Department of Radiation Oncology, University of Maryland, Baltimore, MD (Drs Y. Zhang and X. Zhang); National Technology Institute of Psychiatry, Central South University, Changsha, PR China (Drs Y. Wang and L. Wang); Key Laboratory of Psychiatry and Mental Health of Hunan Province, Central South University, Changsha, PR China (Drs Bai and Zhu)
| | - Mei Bai
- Medical Psychological Institute, Second Xiangya Hospital, Central South University,Changsha, PR China (Drs Y. Zhang, Y. Wang, L. Wang, Bai, and Zhu); Department of Radiation Oncology, University of Maryland, Baltimore, MD (Drs Y. Zhang and X. Zhang); National Technology Institute of Psychiatry, Central South University, Changsha, PR China (Drs Y. Wang and L. Wang); Key Laboratory of Psychiatry and Mental Health of Hunan Province, Central South University, Changsha, PR China (Drs Bai and Zhu)
| | - Xiuwu Zhang
- Medical Psychological Institute, Second Xiangya Hospital, Central South University,Changsha, PR China (Drs Y. Zhang, Y. Wang, L. Wang, Bai, and Zhu); Department of Radiation Oncology, University of Maryland, Baltimore, MD (Drs Y. Zhang and X. Zhang); National Technology Institute of Psychiatry, Central South University, Changsha, PR China (Drs Y. Wang and L. Wang); Key Laboratory of Psychiatry and Mental Health of Hunan Province, Central South University, Changsha, PR China (Drs Bai and Zhu)
| | - Xiongzhao Zhu
- Medical Psychological Institute, Second Xiangya Hospital, Central South University,Changsha, PR China (Drs Y. Zhang, Y. Wang, L. Wang, Bai, and Zhu); Department of Radiation Oncology, University of Maryland, Baltimore, MD (Drs Y. Zhang and X. Zhang); National Technology Institute of Psychiatry, Central South University, Changsha, PR China (Drs Y. Wang and L. Wang); Key Laboratory of Psychiatry and Mental Health of Hunan Province, Central South University, Changsha, PR China (Drs Bai and Zhu).
| |
Collapse
|
29
|
Anderegg A, Awatramani R. Making a mes: A transcription factor-microRNA pair governs the size of the midbrain and the dopaminergic progenitor pool. NEUROGENESIS 2015; 2:e998101. [PMID: 27502145 PMCID: PMC4973584 DOI: 10.1080/23262133.2014.998101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 12/04/2014] [Accepted: 12/10/2014] [Indexed: 02/03/2023]
Abstract
Canonical Wnt signaling is critical for midbrain dopaminergic progenitor specification, proliferation, and neurogenesis. Yet mechanisms that control Wnt signaling remain to be fully elucidated. Wnt1 is a key ligand in the embryonic midbrain, and directs proliferation, survival, specification and neurogenesis. In a recent study, we reveal that the transcription factor Lmx1b promotes Wnt1/Wnt signaling, and dopaminergic progenitor expansion, consistent with earlier studies. Additionally, Lmx1b drives expression of a non-coding RNA called Rmst, which harbors miR135a2 in its last intron. miR135a2 in turn targets Lmx1b as well as several Wnt pathway targets. Conditional overexpression of miR135a2 in the midbrain, particularly during an early time, results in a decreased dopaminergic progenitor pool, and less dopaminergic neurons, consistent with decreased Wnt signaling. We propose a model in which Lmx1b and miR135a2 influence levels of Wnt1 and Wnt signaling, and expansion of the dopaminergic progenitor pool. Further loss of function experiments and biochemical validation of targets will be critical to verify this model. Wnt agonists have recently been utilized for programming stem cells toward a dopaminergic fate in vitro, highlighting the importance of agents that modulate the Wnt pathway.
Collapse
Affiliation(s)
- Angela Anderegg
- Department of Neurology; Northwestern University ; Chicago, IL USA
| | | |
Collapse
|
30
|
Aoi W. Frontier impact of microRNAs in skeletal muscle research: a future perspective. Front Physiol 2015; 5:495. [PMID: 25601837 PMCID: PMC4283715 DOI: 10.3389/fphys.2014.00495] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/01/2014] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are non-coding RNAs that can regulate the expression of mRNAs and proteins by degrading mRNA molecules or by inhibiting their translation. It has been predicted that miRNAs regulate approximately 60% of protein-coding genes that could be involved in a wide range of biological processes. Research over the last 5 years suggests that miRNAs play important roles in skeletal muscle function and several miRNAs have been identified as modulators of myogenesis, muscle mass, and nutrient metabolism in physiological and pathological states. In addition, some miRNAs can be incorporated into intracellular vesicles, released into the circulation, transported to other cells, and possibly function in other organs in an endocrine manner. This phenomenon might explain the interactions between skeletal muscles and other organs. Thus, far, several muscle-secreted miRNAs have been identified and their involvement in muscle biology has been debated. Based on the recent understanding, this perspective article describes the potential valuable role of miRNAs in skeletal muscle function, delineates its limitations, and outlines its future perspectives.
Collapse
Affiliation(s)
- Wataru Aoi
- Laboratory of Health Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University Kyoto, Japan
| |
Collapse
|
31
|
García-Pérez D, López-Bellido R, Hidalgo JM, Rodríguez RE, Laorden ML, Núñez C, Milanés MV. Morphine regulates Argonaute 2 and TH expression and activity but not miR-133b in midbrain dopaminergic neurons. Addict Biol 2015; 20:104-19. [PMID: 23927484 DOI: 10.1111/adb.12083] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Epigenetic changes such as microRNAs (miRs)/Ago2-induced gene silencing represent complex molecular signature that regulate cellular plasticity. Recent studies showed involvement of miRs and Ago2 in drug addiction. In this study, we show that changes in gene expression induced by morphine and morphine withdrawal occur with concomitant epigenetic modifications in the mesolimbic dopaminergic (DA) pathway [ventral tegmental area (VTA)/nucleus accumbens (NAc) shell], which is critically involved in drug-induced dependence. We found that acute or chronic morphine administration as well as morphine withdrawal did not modify miR-133b messenger RNA (mRNA) expression in the VTA, whereas Ago2 protein levels were decreased and increased in morphine-dependent rats and after morphine withdrawal, respectively. These changes were paralleled with enhanced and decreased NAc tyrosine hydroxylase (TH) protein (an early DA marker) in morphine-dependent rats and after withdrawal, respectively. We also observed changes in TH mRNA expression in the VTA that could be related to Ago2-induced translational repression of TH mRNA during morphine withdrawal. However, the VTA number of TH-positive neurons suffered no alterations after the different treatment. Acute morphine administration produced a marked increase in TH activity and DA turnover in the NAc (shell). In contrast, precipitated morphine withdrawal decreased TH activation and did not change DA turnover. These findings provide new information into the possible correlation between Ago2/miRs complex regulation and DA neurons plasticity during opiate addiction.
Collapse
Affiliation(s)
- Daniel García-Pérez
- Group of Cellular and Molecular Pharmacology; Faculty of Medicine; University of Murcia; Murcia Spain
- IMIB (Murcia Institute of Biomedical Investigation); Murcia Spain
| | - Roger López-Bellido
- Department of Biochemistry and Molecular Biology; Institute of Neurosciences; University of Salamanca; Salamanca Spain
| | - Juana M. Hidalgo
- Group of Cellular and Molecular Pharmacology; Faculty of Medicine; University of Murcia; Murcia Spain
- IMIB (Murcia Institute of Biomedical Investigation); Murcia Spain
| | - Raquel E. Rodríguez
- Department of Biochemistry and Molecular Biology; Institute of Neurosciences; University of Salamanca; Salamanca Spain
| | - Maria Luisa Laorden
- Group of Cellular and Molecular Pharmacology; Faculty of Medicine; University of Murcia; Murcia Spain
- IMIB (Murcia Institute of Biomedical Investigation); Murcia Spain
| | - Cristina Núñez
- Group of Cellular and Molecular Pharmacology; Faculty of Medicine; University of Murcia; Murcia Spain
- IMIB (Murcia Institute of Biomedical Investigation); Murcia Spain
| | - Maria Victoria Milanés
- Group of Cellular and Molecular Pharmacology; Faculty of Medicine; University of Murcia; Murcia Spain
- IMIB (Murcia Institute of Biomedical Investigation); Murcia Spain
| |
Collapse
|
32
|
Kirby TJ, Chaillou T, McCarthy JJ. The role of microRNAs in skeletal muscle health and disease. Front Biosci (Landmark Ed) 2015; 20:37-77. [PMID: 25553440 DOI: 10.2741/4298] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the last decade non-coding RNAs have emerged as importance regulators of gene expression. In particular, microRNAs are a class of small RNAs of ∼ 22 nucleotides that repress gene expression through a post-transcriptional mechanism. MicroRNAs have been shown to be involved in a broader range of biological processes, both physiological and pathological, including myogenesis, adaptation to exercise and various myopathies. The purpose of this review is to provide a comprehensive summary of what is currently known about the role of microRNAs in skeletal muscle health and disease.
Collapse
Affiliation(s)
- Tyler J Kirby
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA, 2Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Thomas Chaillou
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA, 2Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - John J McCarthy
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA, 2Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
33
|
Stappert L, Roese-Koerner B, Brüstle O. The role of microRNAs in human neural stem cells, neuronal differentiation and subtype specification. Cell Tissue Res 2015; 359:47-64. [PMID: 25172833 PMCID: PMC4284387 DOI: 10.1007/s00441-014-1981-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 07/28/2014] [Indexed: 12/20/2022]
Abstract
The impressive neuronal diversity found within the nervous system emerges from a limited pool of neural progenitor cells that proceed through different gene expression programs to acquire distinct cell fates. Here, we review recent evidence indicating that microRNAs (miRNAs) are critically involved in conferring neural cell identities during neural induction, neuronal differentiation and subtype specification. Several studies have shown that miRNAs act in concert with other gene regulatory factors and genetic switches to regulate the spatial and temporal expression profiles of important cell fate determinants. So far, most studies addressing the role of miRNAs during neurogenesis were conducted using animal models. With the advent of human pluripotent stem cells and the possibility to differentiate these into neural stem cells, we now have the opportunity to study miRNAs in a human context. More insight into the impact of miRNA-based regulation during neural fate choice could in the end be exploited to develop new strategies for the generation of distinct human neuronal cell types.
Collapse
Affiliation(s)
- Laura Stappert
- Institute of Reconstructive Neurobiology LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Sigmund-Freud-Straße 25, Bonn, 53127 Germany
| | - Beate Roese-Koerner
- Institute of Reconstructive Neurobiology LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Sigmund-Freud-Straße 25, Bonn, 53127 Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Sigmund-Freud-Straße 25, Bonn, 53127 Germany
| |
Collapse
|
34
|
Veenvliet JV, Smidt MP. Molecular mechanisms of dopaminergic subset specification: fundamental aspects and clinical perspectives. Cell Mol Life Sci 2014; 71:4703-27. [PMID: 25064061 PMCID: PMC11113784 DOI: 10.1007/s00018-014-1681-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 07/04/2014] [Accepted: 07/10/2014] [Indexed: 12/22/2022]
Abstract
Dopaminergic (DA) neurons in the ventral mesodiencephalon control locomotion and emotion and are affected in psychiatric and neurodegenerative diseases, such as Parkinson's disease (PD). A clinical hallmark of PD is the specific degeneration of DA neurons located within the substantia nigra (SNc), whereas neurons in the ventral tegmental area remain unaffected. Recent advances have highlighted that the selective vulnerability of the SNc may originate in subset-specific molecular programming during DA neuron development, and significantly increased our understanding of the molecular code that drives specific SNc development. We here present an up-to-date overview of molecular mechanisms that direct DA subset specification, integrating our current knowledge about subset-specific roles of transcription factors, signaling pathways and morphogenes. We discuss strategies to further unravel subset-specific gene-regulatory networks, and the clinical promise of fundamental knowledge about subset specification of DA neurons, with regards to cell replacement therapy and cell-type-specific vulnerability in PD.
Collapse
Affiliation(s)
- Jesse V. Veenvliet
- Department of Molecular Neuroscience, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Marten P. Smidt
- Department of Molecular Neuroscience, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| |
Collapse
|
35
|
The proteomic landscape of the suprachiasmatic nucleus clock reveals large-scale coordination of key biological processes. PLoS Genet 2014; 10:e1004695. [PMID: 25330117 PMCID: PMC4199512 DOI: 10.1371/journal.pgen.1004695] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 08/21/2014] [Indexed: 11/19/2022] Open
Abstract
The suprachiasmatic nucleus (SCN) acts as the central clock to coordinate circadian oscillations in mammalian behavior, physiology and gene expression. Despite our knowledge of the circadian transcriptome of the SCN, how it impacts genome-wide protein expression is not well understood. Here, we interrogated the murine SCN proteome across the circadian cycle using SILAC-based quantitative mass spectrometry. Of the 2112 proteins that were accurately quantified, 20% (421 proteins) displayed a time-of-day-dependent expression profile. Within this time-of-day proteome, 11% (48 proteins) were further defined as circadian based on a sinusoidal expression pattern with a ∼24 h period. Nine circadianly expressed proteins exhibited 24 h rhythms at the transcript level, with an average time lag that exceeded 8 h. A substantial proportion of the time-of-day proteome exhibited abrupt fluctuations at the anticipated light-to-dark and dark-to-light transitions, and was enriched for proteins involved in several key biological pathways, most notably, mitochondrial oxidative phosphorylation. Additionally, predicted targets of miR-133ab were enriched in specific hierarchical clusters and were inversely correlated with miR133ab expression in the SCN. These insights into the proteomic landscape of the SCN will facilitate a more integrative understanding of cellular control within the SCN clock. The suprachiasmatic nucleus (SCN) serves as the master circadian pacemaker in mammals, coordinating the physiological responses of a myriad of peripheral clocks throughout the body and linking their rhythms to the environmental light-dark cycle. In this study, we interrogated the murine SCN proteome across the circadian cycle using stable isotope labeling by amino acids in cell culture (SILAC)-based quantitative mass spectrometry. Among 3275 identified proteins in the SCN, 421 displayed a time-of-day-dependent expression profile, 48 fit a circadian expression profile with a ∼24 h period, and a surprising number of proteins were ultradianly expressed. Nine circadianly expressed proteins were accompanied by transcripts that were also 24 h rhythmic, but with a significant time lag (>8 h) between the phases of peak mRNA vs. protein expression. A substantial proportion of the time-of-day proteome exhibited abrupt fluctuations at the anticipated dawn and dusk, and was involved in mitochondrial oxidative phosphorylation. Additionally, predicted targets of miR-133ab were enriched in specific hierarchical clusters and were inversely correlated with miR133ab expression in the SCN. Our study underscores the significance of post-transcriptional regulation, the surprising prevalence of ultradian protein expression, and the functional implications on mitochondrial energy metabolism.
Collapse
|
36
|
Serafin A, Foco L, Blankenburg H, Picard A, Zanigni S, Zanon A, Pramstaller PP, Hicks AA, Schwienbacher C. Identification of a set of endogenous reference genes for miRNA expression studies in Parkinson's disease blood samples. BMC Res Notes 2014; 7:715. [PMID: 25304816 PMCID: PMC4209045 DOI: 10.1186/1756-0500-7-715] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 10/02/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Research on microRNAs (miRNAs) is becoming an increasingly attractive field, as these small RNA molecules are involved in several physiological functions and diseases. To date, only few studies have assessed the expression of blood miRNAs related to Parkinson's disease (PD) using microarray and quantitative real-time PCR (qRT-PCR). Measuring miRNA expression involves normalization of qRT-PCR data using endogenous reference genes for calibration, but their choice remains a delicate problem with serious impact on the resulting expression levels. The aim of the present study was to evaluate the suitability of a set of commonly used small RNAs as normalizers and to identify which of these miRNAs might be considered reliable reference genes in qRT-PCR expression analyses on PD blood samples. RESULTS Commonly used reference genes snoRNA RNU24, snRNA RNU6B, snoRNA Z30 and miR-103a-3p were selected from the literature. We then analyzed the effect of using these genes as reference, alone or in any possible combination, on the measured expression levels of the target genes miR-30b-5p and miR-29a-3p, which have been previously reported to be deregulated in PD blood samples. CONCLUSIONS We identified RNU24 and Z30 as a reliable and stable pair of reference genes in PD blood samples.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Christine Schwienbacher
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), 39100 Bolzano, Italy, Affiliated Institute of the University of Lübeck, Lübeck, Germany.
| |
Collapse
|
37
|
Zhao N, Jin L, Fei G, Zheng Z, Zhong C. Serum microRNA-133b is associated with low ceruloplasmin levels in Parkinson's disease. Parkinsonism Relat Disord 2014; 20:1177-80. [PMID: 25218846 DOI: 10.1016/j.parkreldis.2014.08.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 08/17/2014] [Accepted: 08/24/2014] [Indexed: 10/24/2022]
Abstract
INTRODUCTION The cause of low serum ceruloplasmin levels in Parkinson's disease (PD) remains to be clarified. In this study, we explored serum miR-133b expression to determine whether it correlates with serum ceruloplasmin level in PD patients. METHODS Forty-six patients with PD and forty-six control subjects were evaluated for miR-133b expression using qRT-PCR. The serum ceruloplasmin levels in all of the subjects were also determined. RESULTS Serum miR-133b expression levels were significantly decreased in PD patients compared with those in the control subjects. Furthermore, PD patients with low serum ceruloplasmin levels also exhibited significantly lower expression of miR-133b compared with that of patients with normal ceruloplasmin levels. MiR-133b expression was correlated with the ceruloplasmin level in patients with PD, whereas no correlation was found between miR-133b and disease severity or motor phenotype. CONCLUSION Our observations suggest that miR-133b might be involved in ceruloplasmin dysmetabolism in PD patients and a further investigation is warranted to confirm this hypothesis.
Collapse
Affiliation(s)
- Na Zhao
- Department of Neurology, Zhongshan Hospital and Shanghai Medical College, Shanghai 200032, China.
| | - Lirong Jin
- Department of Neurology, Zhongshan Hospital and Shanghai Medical College, Shanghai 200032, China.
| | - Guoqiang Fei
- Department of Neurology, Zhongshan Hospital and Shanghai Medical College, Shanghai 200032, China.
| | - Zhiyong Zheng
- Xinxiang College of Medicine, Xinxiang, Henan Province 453003, China.
| | - Chunjiu Zhong
- Department of Neurology, Zhongshan Hospital and Shanghai Medical College, Shanghai 200032, China; Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
38
|
Kim W, Lee Y, McKenna ND, Yi M, Simunovic F, Wang Y, Kong B, Rooney RJ, Seo H, Stephens RM, Sonntag KC. miR-126 contributes to Parkinson's disease by dysregulating the insulin-like growth factor/phosphoinositide 3-kinase signaling. Neurobiol Aging 2014; 35:1712-1721. [PMID: 24559646 PMCID: PMC3991567 DOI: 10.1016/j.neurobiolaging.2014.01.021] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 01/11/2014] [Accepted: 01/19/2014] [Indexed: 01/13/2023]
Abstract
Dopamine (DA) neurons in sporadic Parkinson's disease (PD) display dysregulated gene expression networks and signaling pathways that are implicated in PD pathogenesis. Micro (mi)RNAs are regulators of gene expression, which could be involved in neurodegenerative diseases. We determined the miRNA profiles in laser microdissected DA neurons from postmortem sporadic PD patients' brains and age-matched controls. DA neurons had a distinctive miRNA signature and a set of miRNAs was dysregulated in PD. Bioinformatics analysis provided evidence for correlations of miRNAs with signaling pathways relevant to PD, including an association of miR-126 with insulin/IGF-1/PI3K signaling. In DA neuronal cell systems, enhanced expression of miR-126 impaired IGF-1 signaling and increased vulnerability to the neurotoxin 6-OHDA by downregulating factors in IGF-1/PI3K signaling, including its targets p85β, IRS-1, and SPRED1. Blocking of miR-126 function increased IGF-1 trophism and neuroprotection to 6-OHDA. Our data imply that elevated levels of miR-126 may play a functional role in DA neurons and in PD pathogenesis by downregulating IGF-1/PI3K/AKT signaling and that its inhibition could be a mechanism of neuroprotection.
Collapse
Affiliation(s)
- Woori Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA; Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Yenarae Lee
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Noah D McKenna
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Ming Yi
- Bioinformatics Support Group, Advanced Biomedical Computing Center, NCI-Frederick, Frederick, MD, USA
| | - Filip Simunovic
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | | | | | | | - Hyemyung Seo
- Division of Molecular & Life Sciences, College of Science & Technology, Hanyang University, Seoul, Korea
| | - Robert M Stephens
- Bioinformatics Support Group, Advanced Biomedical Computing Center, NCI-Frederick, Frederick, MD, USA
| | - Kai C Sonntag
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA.
| |
Collapse
|
39
|
Valdez G, Heyer MP, Feng G, Sanes JR. The role of muscle microRNAs in repairing the neuromuscular junction. PLoS One 2014; 9:e93140. [PMID: 24664281 PMCID: PMC3963997 DOI: 10.1371/journal.pone.0093140] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 03/01/2014] [Indexed: 01/11/2023] Open
Abstract
microRNAs have been implicated in mediating key aspects of skeletal muscle development and responses to diseases and injury. Recently, we demonstrated that a synaptically enriched microRNA, miR-206, functions to promote maintenance and repair of the neuromuscular junction (NMJ); in mutant mice lacking miR-206, reinnervation is impaired following nerve injury and loss of NMJs is accelerated in a mouse model of amyotrophic lateral sclerosis (ALS). Here, we asked whether other microRNAs play similar roles. One attractive candidate is miR-133b because it is in the same transcript that encodes miR-206. Like miR-206, miR-133b is concentrated near NMJs and induced after denervation. In miR-133b null mice, however, NMJ development is unaltered, reinnervation proceeds normally following nerve injury, and disease progression is unaffected in the SOD1(G93A) mouse model of ALS. To determine if miR-206 compensates for the loss of miR-133b, we generated mice lacking both microRNAs. The phenotype of these double mutants resembled that of miR-206 single mutants. Finally, we used conditional mutants of Dicer, an enzyme required for the maturation of most microRNAs, to generate mice in which microRNAs were depleted from skeletal muscle fibers postnatally, thus circumventing a requirement for microRNAs in embryonic muscle development. Reinnervation of muscle fibers following injury was impaired in these mice, but the defect was similar in magnitude to that observed in miR-206 mutants. Together, these results suggest that miR-206 is the major microRNA that regulates repair of the NMJ following nerve injury.
Collapse
Affiliation(s)
- Gregorio Valdez
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States of America
- Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke, Virginia, United States of America
- * E-mail:
| | - Mary P. Heyer
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Molecular Therapeutics, The Scripps Research Institute Florida, Jupiter, Florida, United States of America
| | - Guoping Feng
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, United States of America
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Joshua R. Sanes
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States of America
| |
Collapse
|
40
|
Schlaudraff F, Gründemann J, Fauler M, Dragicevic E, Hardy J, Liss B. Orchestrated increase of dopamine and PARK mRNAs but not miR-133b in dopamine neurons in Parkinson's disease. Neurobiol Aging 2014; 35:2302-15. [PMID: 24742361 PMCID: PMC4099518 DOI: 10.1016/j.neurobiolaging.2014.03.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 02/27/2014] [Accepted: 03/14/2014] [Indexed: 01/25/2023]
Abstract
Progressive loss of substantia nigra dopamine neurons (SN DA) is a hallmark of aging and of Parkinson's disease (PD). Mutations in PARK genes cause familial PD forms. Increased expression of alpha-synuclein (PARK4) is a disease-triggering event in familial PD and also observed in SN DA neurons in sporadic PD but related transcriptional changes are unknown. With optimized single-cell quantitative real-time polymerase chain reaction analysis, we compared messenger RNA and microRNA levels in SN DA neurons from sporadic PD patients and controls. Non-optimally matched donor ages and RNA integrities are common problems when analyzing human samples. We dissected the influence of distinct ages and RNA integrities of our samples by applying a specifically-optimized, linear-mixed-effects model to quantitative real-time polymerase chain reaction-data. We identified that elevated alpha-synuclein messenger RNA levels in SN DA neurons of human PD brains were positively correlated with corresponding elevated levels of mRNAs for functional compensation of progressive SN DA loss and for enhanced proteasomal (PARK5/UCHL1) and lysosomal (PARK9/ATPase13A2) function, possibly counteracting alpha-synuclein toxicity. In contrast, microRNA miR-133b levels, previously implicated in transcriptional dysregulation in PD, were not altered in SN DA neurons in PD.
Collapse
Affiliation(s)
- Falk Schlaudraff
- Department of Applied Physiology, Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | - Jan Gründemann
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Michael Fauler
- Department of Applied Physiology, Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | - Elena Dragicevic
- Department of Applied Physiology, Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | - John Hardy
- Department of Molecular Neuroscience and Reta Lila Weston Laboratories, Institute of Neurology, London, UK
| | - Birgit Liss
- Department of Applied Physiology, Institute of Applied Physiology, University of Ulm, Ulm, Germany.
| |
Collapse
|
41
|
Follert P, Cremer H, Béclin C. MicroRNAs in brain development and function: a matter of flexibility and stability. Front Mol Neurosci 2014; 7:5. [PMID: 24570654 PMCID: PMC3916726 DOI: 10.3389/fnmol.2014.00005] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 01/16/2014] [Indexed: 01/27/2023] Open
Abstract
Fine-tuning of gene expression is a fundamental requirement for development and function of cells and organs. This requirement is particularly obvious in the nervous system where originally common stem cell populations generate thousands of different neuronal and glial cell types in a temporally and quantitatively perfectly orchestrated manner. Moreover, after their generation, young neurons have to connect with pre-determined target neurons through the establishment of functional synapses, either in their immediate environment or at distance. Lastly, brain function depends not only on static circuitries, but on plastic changes at the synaptic level allowing both, learning and memory. It appears evident that these processes necessitate flexibility and stability at the same time. These two contrasting features can only be achieved by complex molecular networks, superposed levels of control and tight interactions between regulatory mechanisms. Interactions between microRNAs and their target mRNAs fulfill these requirements. Here we review recent literature dealing with the involvement of microRNAs in multiple aspects of brain development and connectivity.
Collapse
Affiliation(s)
- Philipp Follert
- Institut de Biologie du Développement de Marseille, Aix-Marseille Université - Centre National de la Recherche Scientifique Marseille, France
| | - Harold Cremer
- Institut de Biologie du Développement de Marseille, Aix-Marseille Université - Centre National de la Recherche Scientifique Marseille, France
| | - Christophe Béclin
- Institut de Biologie du Développement de Marseille, Aix-Marseille Université - Centre National de la Recherche Scientifique Marseille, France
| |
Collapse
|
42
|
Higa GSV, de Sousa E, Walter LT, Kinjo ER, Resende RR, Kihara AH. MicroRNAs in neuronal communication. Mol Neurobiol 2014; 49:1309-26. [PMID: 24385256 DOI: 10.1007/s12035-013-8603-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 12/05/2013] [Indexed: 12/28/2022]
Abstract
MicroRNAs (miRNAs) are short nucleotides sequences that regulate the expression of genes in different eukaryotic cell types. A tremendous amount of knowledge on miRNAs has rapidly accumulated over the last few years, revealing the growing interest in this field of research. On the other hand, clarifying the physiological regulation of gene expression in the central nervous system is important for establishing a reference for comparison to the diseased state. It is well known that the fine tuning of neuronal networks relies on intricate molecular mechanisms, such as the adjustment of the synaptic transmission. As determined by recent studies, regulation of neuronal interactions by miRNAs has critical consequences in the development, adaptation to ambient demands, and degeneration of the nervous system. In contrast, activation of synaptic receptors triggers downstream signaling cascades that generate a vast array of effects, which includes the regulation of novel genes involved in the control of the miRNA life cycle. In this review, we have examined the hot topics on miRNA gene-regulatory activities in the broad field of neuronal communication-related processes. Furthermore, in addition to indicating the newly described effect of miRNAs on the regulation of specific neurotransmitter systems, we have pointed out how these systems affect the expression, transport, and stability of miRNAs. Moreover, we discuss newly described and under-investigation mechanisms involving the intercellular transfer of miRNAs, aided by exosomes and gap junctions. Thus, in the current review, we were able to highlight recent findings related to miRNAs that indisputably contributed towards the understanding of the nervous system in health and disease.
Collapse
Affiliation(s)
- Guilherme Shigueto Vilar Higa
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Av. Atlântica 420, 09060-000, Santo André, SP, Brazil
| | | | | | | | | | | |
Collapse
|
43
|
Heman-Ackah SM, Hallegger M, Rao MS, Wood MJA. RISC in PD: the impact of microRNAs in Parkinson's disease cellular and molecular pathogenesis. Front Mol Neurosci 2013; 6:40. [PMID: 24312000 PMCID: PMC3834244 DOI: 10.3389/fnmol.2013.00040] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/31/2013] [Indexed: 12/19/2022] Open
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative disease characterized primarily by the selective death of dopaminergic (DA) neurons in the substantia nigra pars compacta of the midbrain. Although several genetic forms of PD have been identified, the precise molecular mechanisms underlying DA neuron loss in PD remain elusive. In recent years, microRNAs (miRNAs) have been recognized as potent post-transcriptional regulators of gene expression with fundamental roles in numerous biological processes. Although their role in PD pathogenesis is still a very active area of investigation, several seminal studies have contributed significantly to our understanding of the roles these small non-coding RNAs play in the disease process. Among these are studies which have demonstrated specific miRNAs that target and down-regulate the expression of PD-related genes as well as those demonstrating a reciprocal relationship in which PD-related genes act to regulate miRNA processing machinery. Concurrently, a wealth of knowledge has become available regarding the molecular mechanisms that unify the underlying etiology of genetic and sporadic PD pathogenesis, including dysregulated protein quality control by the ubiquitin-proteasome system and autophagy pathway, activation of programmed cell death, mitochondrial damage and aberrant DA neurodevelopment and maintenance. Following a discussion of the interactions between PD-related genes and miRNAs, this review highlights those studies which have elucidated the roles of these pathways in PD pathogenesis. We highlight the potential of miRNAs to serve a critical regulatory role in the implicated disease pathways, given their capacity to modulate the expression of entire families of related genes. Although few studies have directly linked miRNA regulation of these pathways to PD, a strong foundation for investigation has been laid and this area holds promise to reveal novel therapeutic targets for PD.
Collapse
Affiliation(s)
- Sabrina M Heman-Ackah
- Department of Physiology, Anatomy and Genetics, University of Oxford Oxford, UK ; Center for Regenerative Medicine, US National Institutes of Health Bethesda, MD, USA
| | | | | | | |
Collapse
|
44
|
Dicer expression is essential for adult midbrain dopaminergic neuron maintenance and survival. Mol Cell Neurosci 2013; 58:22-8. [PMID: 24184162 DOI: 10.1016/j.mcn.2013.10.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 08/20/2013] [Accepted: 10/23/2013] [Indexed: 01/28/2023] Open
Abstract
The type III RNAse, Dicer, is responsible for the processing of microRNA (miRNA) precursors into functional miRNA molecules, non-coding RNAs that bind to and target messenger RNAs for repression. Dicer expression is essential for mouse midbrain development and dopaminergic (DAergic) neuron maintenance and survival during the early post-natal period. However, the role of Dicer in adult mouse DAergic neuron maintenance and survival is unknown. To bridge this gap in knowledge, we selectively knocked-down Dicer expression in individual DAergic midbrain areas, including the ventral tegmental area (VTA) and substantia nigra pars compacta (SNpc) via viral-mediated expression of Cre in adult floxed Dicer knock-in mice (Dicer(flox/flox)). Bilateral Dicer loss in the VTA resulted in progressive hyperactivity that was significantly reduced by the dopamine agonist, amphetamine. In contrast, decreased Dicer expression in the SNpc did not affect locomotor activity but did induce motor-learning impairment on an accelerating rotarod. Knock-down of Dicer in both midbrain regions of adult Dicer(flox/flox) mice resulted in preferential, progressive loss of DAergic neurons likely explaining motor behavior phenotypes. In addition, knock-down of Dicer in midbrain areas triggered neuronal death via apoptosis. Together, these data indicate that Dicer expression and, as a consequence, miRNA function, are essential for DAergic neuronal maintenance and survival in adult midbrain DAergic neuron brain areas.
Collapse
|
45
|
Goodall EF, Heath PR, Bandmann O, Kirby J, Shaw PJ. Neuronal dark matter: the emerging role of microRNAs in neurodegeneration. Front Cell Neurosci 2013; 7:178. [PMID: 24133413 PMCID: PMC3794211 DOI: 10.3389/fncel.2013.00178] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 09/21/2013] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs (miRNAs) are small, abundant RNA molecules that constitute part of the cell's non-coding RNA “dark matter.” In recent years, the discovery of miRNAs has revolutionised the traditional view of gene expression and our understanding of miRNA biogenesis and function has expanded. Altered expression of miRNAs is increasingly recognized as a feature of many disease states, including neurodegeneration. Here, we review the emerging role for miRNA dysfunction in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis (ALS) and Huntington's disease pathogenesis. We emphasize the complex nature of gene regulatory networks and the need for systematic studies, with larger sample cohorts than have so far been reported, to reveal the most important miRNA regulators in disease. Finally, miRNA diversity and their potential to target multiple pathways, offers novel clinical applications for miRNAs as biomarkers and therapeutic agents in neurodegenerative diseases.
Collapse
Affiliation(s)
- Emily F Goodall
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield Sheffield, UK
| | | | | | | | | |
Collapse
|
46
|
Khoo SK, Neuman LA, Forsgren L, Petillo D, Brundin P. Could miRNA expression changes be a reliable clinical biomarker for Parkinson’s disease? Neurodegener Dis Manag 2013. [DOI: 10.2217/nmt.13.53] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
SUMMARY Parkinson’s disease (PD) is a complex neurological disorder involving progressive loss of dopaminergic and other neurons, as well as intraneuronal aggregation of α-synuclein. These changes lead to motor, cognitive and multiple non-motor clinical issues. Currently, PD diagnosis is primarily based on its motor symptoms, which are delayed and subtle; neurodegeneration is believed to reach a relatively advanced stage by the time of clinical diagnosis. Developing reliable clinical biomarkers that are objective, measurable, specific and sensitive for early detection of PD remains a challenge. miRNAs are small, noncoding RNAs involved in development and gene regulation. miRNAs possess unique and ideal biomarker characteristics: highly abundant, stable and quantifiable. Here, we review a list of miRNA candidates that could potentially represent clinical biomarkers for PD.
Collapse
Affiliation(s)
- Sok Kean Khoo
- Genomic Microarray Core, Van Andel Institute, Grand Rapids, MI, USA
- Department of Cell & Molecular Biology, Grand Valley State University, Grand Rapids, MI, USA
| | - Leslie A Neuman
- Neuroscience Program, Mercy Health Saint Mary’s, Grand Rapids, MI, USA
| | - Lars Forsgren
- Department of Pharmacology & Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - David Petillo
- Center for Cancer & Cell Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| |
Collapse
|
47
|
Kirby TJ, McCarthy JJ. MicroRNAs in skeletal muscle biology and exercise adaptation. Free Radic Biol Med 2013; 64:95-105. [PMID: 23872025 PMCID: PMC4867469 DOI: 10.1016/j.freeradbiomed.2013.07.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 07/01/2013] [Accepted: 07/02/2013] [Indexed: 01/01/2023]
Abstract
MicroRNAs (miRNAs) have emerged as important players in the regulation of gene expression, being involved in most biological processes examined to date. The proposal that miRNAs are primarily involved in the stress response of the cell makes miRNAs ideally suited to mediate the response of skeletal muscle to changes in contractile activity. Although the field is still in its infancy, the studies presented in this review highlight the promise that miRNAs will have an important role in mediating the response and adaptation of skeletal muscle to various modes of exercise. The roles of miRNAs in satellite cell biology, muscle regeneration, and various myopathies are also discussed.
Collapse
Affiliation(s)
- Tyler J. Kirby
- Department of Physiology, University of Kentucky Lexington, KY 40516-0298
| | - John J. McCarthy
- Department of Physiology, University of Kentucky Lexington, KY 40516-0298
- Center for Muscle Biology, University of Kentucky Lexington, KY 40516-0298
| |
Collapse
|
48
|
Dissecting the diversity of midbrain dopamine neurons. Trends Neurosci 2013; 36:336-42. [DOI: 10.1016/j.tins.2013.03.003] [Citation(s) in RCA: 261] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 03/11/2013] [Accepted: 03/14/2013] [Indexed: 11/17/2022]
|
49
|
Bicchi I, Morena F, Montesano S, Polidoro M, Martino S. MicroRNAs and Molecular Mechanisms of Neurodegeneration. Genes (Basel) 2013; 4:244-63. [PMID: 24705162 PMCID: PMC3899972 DOI: 10.3390/genes4020244] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 05/09/2013] [Accepted: 05/10/2013] [Indexed: 12/21/2022] Open
Abstract
During the last few years microRNAs (miRNAs) have emerged as key mediators of post-transcriptional and epigenetic regulation of gene expression. MiRNAs targets, identified through gene expression profiling and studies in animal models, depict a scenario where miRNAs are fine-tuning metabolic pathways and genetic networks in both plants and animals. MiRNAs have shown to be differentially expressed in brain areas and alterations of miRNAs homeostasis have been recently correlated to pathological conditions of the nervous system, such as cancer and neurodegeneration. Here, we review and discuss the most recent insights into the involvement of miRNAs in the neurodegenerative mechanisms and their correlation with significant neurodegenerative disorders.
Collapse
Affiliation(s)
- Ilaria Bicchi
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Francesco Morena
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Simona Montesano
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Mario Polidoro
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Sabata Martino
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| |
Collapse
|
50
|
Stappert L, Borghese L, Roese-Koerner B, Weinhold S, Koch P, Terstegge S, Uhrberg M, Wernet P, Brüstle O. MicroRNA-based promotion of human neuronal differentiation and subtype specification. PLoS One 2013; 8:e59011. [PMID: 23527072 PMCID: PMC3601127 DOI: 10.1371/journal.pone.0059011] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 01/17/2013] [Indexed: 11/19/2022] Open
Abstract
MicroRNAs are key regulators of neural cell proliferation, differentiation and fate choice. Due to the limited access to human primary neural tissue, the role of microRNAs in human neuronal differentiation remains largely unknown. Here, we use a population of long-term self-renewing neuroepithelial-like stem cells (lt-NES cells) derived from human embryonic stem cells to study the expression and function of microRNAs at early stages of human neural stem cell differentiation and neuronal lineage decision. Based on microRNA expression profiling followed by gain- and loss-of-function analyses in lt-NES cells and their neuronal progeny, we demonstrate that miR-153, miR-324-5p/3p and miR-181a/a contribute to the shift of lt-NES cells from self-renewal to neuronal differentiation. We further show that miR-125b and miR-181a specifically promote the generation of neurons of dopaminergic fate, whereas miR-181a inhibits the development of this neurotransmitter subtype. Our data demonstrate that time-controlled modulation of specific microRNA activities not only regulates human neural stem cell self-renewal and differentiation but also contributes to the development of defined neuronal subtypes.
Collapse
Affiliation(s)
- Laura Stappert
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Bonn, Germany
| | - Lodovica Borghese
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Bonn, Germany
- * E-mail: (LB); (OB)
| | - Beate Roese-Koerner
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Bonn, Germany
| | - Sandra Weinhold
- Institute for Transplantation Diagnostics and Cell Therapeutics, University Düsseldorf, Düsseldorf, Germany
| | - Philipp Koch
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Bonn, Germany
| | - Stefanie Terstegge
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Bonn, Germany
| | - Markus Uhrberg
- Institute for Transplantation Diagnostics and Cell Therapeutics, University Düsseldorf, Düsseldorf, Germany
| | - Peter Wernet
- Institute for Transplantation Diagnostics and Cell Therapeutics, University Düsseldorf, Düsseldorf, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Bonn, Germany
- * E-mail: (LB); (OB)
| |
Collapse
|